US10570418B2 - Methods and compositions for RNA-directed target DNA modification - Google Patents

Methods and compositions for RNA-directed target DNA modification Download PDF

Info

Publication number
US10570418B2
US10570418B2 US15/502,720 US201515502720A US10570418B2 US 10570418 B2 US10570418 B2 US 10570418B2 US 201515502720 A US201515502720 A US 201515502720A US 10570418 B2 US10570418 B2 US 10570418B2
Authority
US
United States
Prior art keywords
rna
cell
dna
protein
cas9
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US15/502,720
Other languages
English (en)
Other versions
US20180044700A1 (en
Inventor
Jennifer A. DOUDNA
Steven Lin
Brett T. STAAHL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Priority to US15/502,720 priority Critical patent/US10570418B2/en
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STAAHL, Brett T., DOUDNA, JENNIFER A., LIN, STEVEN
Publication of US20180044700A1 publication Critical patent/US20180044700A1/en
Application granted granted Critical
Publication of US10570418B2 publication Critical patent/US10570418B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • C12P19/30Nucleotides
    • C12P19/34Polynucleotides, e.g. nucleic acids, oligoribonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3233Morpholino-type ring
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • dsDNA site-specific double-strand DNA breaks
  • DSBs double-strand DNA breaks
  • ZFNs Zinc-finger nucleases
  • TALENs transcription activator-like effector nucleases
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • Cas9 CRISPR-associated loci encode RNA-guided adaptive immune systems that can destroy foreign DNA.
  • the Type II CRISPR/Cas systems require a single protein, Cas9, to catalyze DNA cleavage.
  • Cas9 generates blunt DSBs at sites defined by a guide sequence contained within an associated CRISPR RNA (crRNA) transcript.
  • Cas9 can modify (e.g., cleave) double-stranded DNA at any site: (i) defined by the guide RNA sequence, and (ii) including a protospacer-adjacent (PAM) motif.
  • a Cas9/guide RNA complex i.e., a Cas9 targeting complex
  • a Cas9 targeting complex having a mutated Cas9 protein with reduced or removed nuclease activity can still bind to target DNA.
  • the present disclosure provides methods and compositions for site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell.
  • the present disclosure provides methods of binding a target DNA in a eukaryotic cell.
  • Subject methods of site-specific modification and/or subject methods of binding can include a step of enriching a cell population for cells that are in a desired phase(s) of the cell cycle (e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.); and a step of contacting the target DNA of cells of the enriched population of cells (e.g., in some cases the target DNA in cells of the enriched population of cells) with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex).
  • a desired phase(s) of the cell cycle e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.
  • a step of contacting the target DNA of cells of the enriched population of cells e.g., in some cases the target DNA in
  • the subject methods include contacting the target DNA in the target cell(s) with: (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein.
  • a step of enriching can include, for example, a cell separation method (e.g., mitotic shake-off, countercurrent centrifugal elutriation (CCE), flow cytometry, and the like) and/or a cell synchronization method (e.g., contact with a cell cycle blocking composition; mitogen and/or growth factor withdrawal; density arrest; and the like).
  • a cell separation method e.g., mitotic shake-off, countercurrent centrifugal elutriation (CCE), flow cytometry, and the like
  • a cell synchronization method e.g., contact with a cell cycle blocking composition; mitogen and/or growth factor withdrawal; density arrest; and the
  • a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at a desired phase in the cell cycle (e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.); and a step of contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex).
  • blocking a cell at a phase in the cell cycle includes contacting the cell with a cell cycle blocking agent.
  • the above methods include a step of contacting a eukaryotic cell, or contacting a population of eukaryotic cells, with a cell cycle blocking composition.
  • a cell cycle blocking composition for use in the above methods includes at least one agent selected from: nocodazole, hydroxyurea; colchicine; demecolcine (colcemid); lovastatin; mimosine; thymidine; aphidicolin; latrunculin A; and latrunculin.
  • the above methods include contacting the target DNA with a Cas9 targeting complex, which in some cases includes introducing into the cell (i.e., the target cell having the target DNA) at least one of: (a) a single guide RNA, (b) a DNA polynucleotide encoding a single guide RNA, (c) a targeter-RNA, (d) a DNA polynucleotide encoding a targeter-RNA, (e) an activator-RNA, (f) a DNA polynucleotide encoding an activator-RNA, (g) a Cas9 protein, and (h) a nucleic acid encoding a Cas9 protein.
  • a Cas9 targeting complex which in some cases includes introducing into the cell (i.e., the target cell having the target DNA) at least one of: (a) a single guide RNA, (b) a DNA polynucleotide encoding a single guide RNA, (c) a targeter-RNA
  • the Cas9 protein has nuclease activity and the site-specific modification is cleavage of the target DNA.
  • the target DNA is double stranded and the Cas9 protein: (a) cleaves the complementary strand of the target DNA, but does not cleave the non-complementary strand of the target DNA; (b) cleaves the non-complementary strand of the target DNA, but does not cleave the complementary strand of the target DNA; (c) cleaves both the complementary and non-complementary strands of the target DNA.
  • the above methods include contacting the target DNA with a donor polynucleotide.
  • the method is a method of binding a target DNA and the Cas9 protein does not cleave the target DNA.
  • the method is a method of site-specific modification of a target DNA and the Cas9 protein has a heterologous polypeptide sequence that provides for a DNA modifying activity (e.g., methyltransferase activity, demethylase activity, DNA repair activity, DNA damage activity, deamination activity, dismutase activity, alkylation activity, depurination activity, oxidation activity, pyrimidine dimer forming activity, integrase activity, transposase activity, recombinase activity, polymerase activity, ligase activity, helicase activity, photolyase activity, and/or glycosylase activity.)
  • the method is a method of site-specific modification of a protein associated with a target DNA
  • the Cas9 protein has a heterologous polypeptide sequence that provides for a protein modifying activity (e.g., methyltransferase activity, demethylase activity, acetyltransferase activity, de
  • the method is a method of binding target DNA in a eukaryotic cell, and the method includes: (a) blocking the cell at a desired phase in the cell cycle; and (b) contacting the target DNA in the cell with: (i) a Cas9 protein, wherein the Cas9 protein does not cleave the target DNA, and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding domain that interacts with the Cas9 protein.
  • the Cas9 protein includes a fusion partner having at least one of: a subcellular localization sequence (e.g., a nuclear localization signal), and a detectable label (e.g., a fluorescent tag, an affinity tag, and the like).
  • a subcellular localization sequence e.g., a nuclear localization signal
  • a detectable label e.g., a fluorescent tag, an affinity tag, and the like.
  • the method is a method of binding target DNA in cells of a population of eukaryotic cells, and the method includes: (a) enriching the population of eukaryotic cells for cells in a desired phase of the cell cycle; and (b) contacting the target DNA of cells of the enriched population of cells (e.g., in some cases the target DNA in cells of the enriched population of cells) with: (i) a Cas9 protein, wherein the Cas9 protein does not cleave the target DNA, and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding domain that interacts with the Cas9 protein.
  • the Cas9 protein includes a fusion partner having at least one of: a subcellular localization sequence (e.g., a nuclear localization signal), and a detectable label (e.g., a fluorescent tag, an affinity tag, and the like).
  • the Cas9 protein includes a fusion partner having at least one of: a subcellular localization sequence (e.g., a nuclear localization signal), and a detectable label (e.g., a fluorescent tag, an affinity tag, and the like).
  • any of the above methods are carried out in a living cell in vitro, in a living cell ex vivo, or in a living cell in vivo.
  • the target cell (or the population of target cells) in any of the above methods is a eukaryotic single-cell organism, a somatic cell, a germ cell, a stem cell, a plant cell, an algal cell, an animal cell, in invertebrate cell, a vertebrate cell, a fish cell, a frog cell, a bird cell, a mammalian cell, a pig cell, a cow cell, a goat cell, a sheep cell, a rodent cell, a rat cell, a mouse cell, a non-human primate cell, or a human cell.
  • Kits and compositions for carrying out the methods are also provided.
  • a subject kit includes a cell cycle blocking agent and at least one component of a Cas9 targeting complex (or a nucleic acid encoding at least one component of a Cas9 targeting complex).
  • FIG. 1A-1D depict the effect of cell cycle synchronization on NHEJ and HDR frequencies in HEK293T cells.
  • FIG. 2A-2B depict the enhancement of NHEJ and HDR at the EMX1 and DYRK1 loci by nocodazole synchronization.
  • FIG. 3A-3B depict systematic investigation of DNA templates for efficient HDR at the EMX1 locus.
  • FIG. 3A depicts nucleotide sequences of a double stranded target (GCCAATGGGGAGGACATCGATGTCACCTCCAATGACTAGGGTGGGCAAC (SEQ ID NO:1408; and its complement GTTGCCCACCCTAGTCATTGGAGGTGACATCGATGTCCTCCCCATTGGC (SEQ ID NO:1409), and various HDR templates (template 1: CCACCGCTACGAAGCTTGTCATTGGAGGTGACATCGATGTCCTC (SEQ ID NO:1410); template 2: GGATCCAAGCTT (SEQ ID NO:1411); template 3: GGATCCAAGCTT (SEQ ID NO:1412); template 4: GGATCCAAGCTT (SEQ ID NO:1413); template 5: AAGCTTGGATCC (SEQ ID NO:1414); template 6: AAGCTTGGATCC (SEQ ID NO:1415) and
  • FIG. 4A-4B depict NHEJ and HDR efficiencies at EMX1 locus in human primary neonatal fibroblast and embryonic stem cells.
  • FIG. 5A-5B provide a schematic drawing of a Cas9 targeting complex binding to a target sequence of a target DNA.
  • Panels A and B depict two exemplary guide RNAs (A-dual guide RNA; B-single guide RNA), each associated with a Cas9 protein and with a target DNA.
  • FIG. 6A-6F provide data related to enhanced gene targeting by cell synchronization and co-delivery of AAV and Cas9 ribonucleoprotein (RNP).
  • FIG. 6B top sequence (SEQ ID NO:1419) and bottom sequence (SEQ ID NO:1420).
  • polynucleotide and “nucleic acid,” used interchangeably herein, refer to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. Thus, this term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases.
  • Oligonucleotide generally refers to polynucleotides of between about 5 and about 100 nucleotides of single- or double-stranded DNA.
  • oligonucleotide is also known as “oligomers” or “oligos” and may be isolated from genes, or chemically synthesized by methods known in the art.
  • polynucleotide and nucleic acid should be understood to include, as applicable to the embodiments being described, single-stranded (such as sense or antisense) and double-stranded polynucleotides.
  • a “stem-loop structure” refers to a nucleic acid having a secondary structure that includes a region of nucleotides which are known or predicted to form a double strand (step portion) that is linked on one side by a region of predominantly single-stranded nucleotides (loop portion).
  • the terms “hairpin” and “fold-back” structures are also used herein to refer to stem-loop structures. Such structures are well known in the art and these terms are used consistently with their known meanings in the art.
  • a stem-loop structure does not require exact base-pairing.
  • the stem may include one or more base mismatches.
  • the base-pairing may be exact, i.e. not include any mismatches.
  • hybridizable or “complementary” or “substantially complementary” it is meant that a nucleic acid (e.g. RNA) comprises a sequence of nucleotides that enables it to non-covalently bind, i.e. form Watson-Crick base pairs and/or G/U base pairs, “anneal”, or “hybridize,” to another nucleic acid in a sequence-specific, antiparallel, manner (i.e., a nucleic acid specifically binds to a complementary nucleic acid) under the appropriate in vitro and/or in vivo conditions of temperature and solution ionic strength.
  • RNA complementary nucleic acid
  • standard Watson-Crick base-pairing includes: adenine (A) pairing with thymidine (T), adenine (A) pairing with uracil (U), and guanine (G) pairing with cytosine (C) [DNA, RNA].
  • A adenine
  • U uracil
  • G guanine
  • C cytosine
  • G/U base-pairing is partially responsible for the degeneracy (i.e., redundancy) of the genetic code in the context of tRNA anti-codon base-pairing with codons in mRNA.
  • a guanine (G) of a protein-binding segment (dsRNA duplex) of a subject guide RNA molecule is considered complementary to a uracil (U), and vice versa.
  • G guanine
  • U uracil
  • Hybridization and washing conditions are well known and exemplified in Sambrook, J., Fritsch, E. F. and Maniatis, T. Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1989), particularly Chapter 11 and Table 11.1 therein; and Sambrook, J. and Russell, W., Molecular Cloning: A Laboratory Manual, Third Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (2001).
  • the conditions of temperature and ionic strength determine the “stringency” of the hybridization.
  • Hybridization requires that the two nucleic acids contain complementary sequences, although mismatches between bases are possible.
  • the conditions appropriate for hybridization between two nucleic acids depend on the length of the nucleic acids and the degree of complementation, variables well known in the art. The greater the degree of complementation between two nucleotide sequences, the greater the value of the melting temperature (Tm) for hybrids of nucleic acids having those sequences.
  • Tm melting temperature
  • For hybridizations between nucleic acids with short stretches of complementarity e.g. complementarity over 35 or less, 30 or less, 25 or less, 22 or less, 20 or less, or 18 or less nucleotides
  • the position of mismatches becomes important (see Sambrook et al., supra, 11.7-11.8).
  • the length for a hybridizable nucleic acid is at least about 10 nucleotides.
  • Illustrative minimum lengths for a hybridizable nucleic acid are: at least about 15 nucleotides; at least about 20 nucleotides; at least about 22 nucleotides; at least about 25 nucleotides; and at least about 30 nucleotides).
  • the temperature and wash solution salt concentration may be adjusted as necessary according to factors such as length of the region of complementation and the degree of complementation.
  • polynucleotide need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable or hybridizable. Moreover, a polynucleotide may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a loop structure or hairpin structure).
  • a polynucleotide can comprise at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or 100% sequence complementarity to a target region within the target nucleic acid sequence to which they are targeted.
  • an antisense nucleic acid in which 18 of 20 nucleotides of the antisense compound are complementary to a target region, and would therefore specifically hybridize would represent 90 percent complementarity.
  • the remaining noncomplementary nucleotides may be clustered or interspersed with complementary nucleotides and need not be contiguous to each other or to complementary nucleotides. Percent complementarity between particular stretches of nucleic acid sequences within nucleic acids can be determined routinely using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. Mol.
  • peptide refers to a polymeric form of amino acids of any length, which can include coded and non-coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones.
  • Binding refers to a non-covalent interaction between macromolecules (e.g., between a protein and a nucleic acid). While in a state of non-covalent interaction, the macromolecules are said to be “associated” or “interacting” or “binding” (e.g., when a molecule X is said to interact with a molecule Y, it is meant the molecule X binds to molecule Y in a non-covalent manner).
  • Binding interactions are generally characterized by a dissociation constant (Kd) of less than 10 ⁇ 6 M, less than 10 ⁇ 7 M, less than 10 ⁇ 8 M, less than 10 ⁇ 9 M, less than 10 ⁇ 10 M, less than 10 ⁇ 11 M, less than 10 ⁇ 12 M, less than 10 ⁇ 13 M, less than 10 ⁇ 14 M, or less than 10 ⁇ 15 M.
  • Kd dissociation constant
  • Affinity refers to the strength of binding, increased binding affinity being correlated with a lower Kd.
  • binding domain it is meant a protein domain that is able to bind non-covalently to another molecule.
  • a binding domain can bind to, for example, a DNA molecule (a DNA-binding protein), an RNA molecule (an RNA-binding protein) and/or a protein molecule (a protein-binding protein).
  • a protein domain-binding protein it can bind to itself (to form homodimers, homotrimers, etc.) and/or it can bind to one or more molecules of a different protein or proteins.
  • a group of amino acids having aliphatic side chains consists of glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains consists of serine and threonine; a group of amino acids having amide containing side chains consisting of asparagine and glutamine; a group of amino acids having aromatic side chains consists of phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains consists of lysine, arginine, and histidine; a group of amino acids having acidic side chains consists of glutamate and aspartate; and a group of amino acids having sulfur containing side chains consists of cysteine and methionine.
  • Exemplary conservative amino acid substitution groups are: valine-leucine-isoleucine, phenyla
  • a polynucleotide or polypeptide has a certain percent “sequence identity” to another polynucleotide or polypeptide, meaning that, when aligned, that percentage of bases or amino acids are the same, and in the same relative position, when comparing the two sequences. Sequence identity can be determined in a number of different manners.
  • sequences can be aligned using various methods and computer programs (e.g., BLAST, T-COFFEE, MUSCLE, MAFFT, etc.), available over the world wide web at sites including ncbi.nlm.nili.gov/BLAST, ebi.ac.uk/Tools/msa/tcoffee/, ebi.ac.uk/Tools/msa/muscle/, mafft.cbrc.jp/alignment/software/. See, e.g., Altschul et al. (1990), J. Mol. Bioi. 215:403-10.
  • a DNA sequence that “encodes” a particular RNA is a DNA nucleic acid sequence that is transcribed into RNA.
  • a DNA polynucleotide may encode an RNA (mRNA) that is translated into protein, or a DNA polynucleotide may encode an RNA that is not translated into protein (e.g. tRNA, rRNA, or a guide RNA; also called “non-coding” RNA or “ncRNA”).
  • a “protein coding sequence” or a sequence that encodes a particular protein is a nucleic acid sequence that is transcribed into mRNA (in the case of DNA) and is translated (in the case of mRNA) into a polypeptide in vitro or in vivo when placed under the control of appropriate regulatory sequences.
  • the boundaries of the coding sequence are determined by a start codon at the 5′ terminus (N-terminus) and a translation stop nonsense codon at the 3′ terminus (C-terminus).
  • a coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and synthetic nucleic acids.
  • a transcription termination sequence will usually be located 3′ to the coding sequence.
  • a “promoter sequence” is a DNA regulatory region capable of binding RNA polymerase and initiating transcription of a downstream (3′ direction) coding or non-coding sequence.
  • the promoter sequence is bounded at its 3′ terminus by the transcription initiation site and extends upstream (5′ direction) to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background.
  • a transcription initiation site within the promoter sequence will be found a transcription initiation site, as well as protein binding domains responsible for the binding of RNA polymerase.
  • Eukaryotic promoters will often, but not always, contain “TATA” boxes and “CAT” boxes.
  • Various promoters, including inducible promoters may be used to drive the various vectors of the present invention.
  • a promoter can be a constitutively active promoter (i.e., a promoter that is constitutively in an active/“ON” state), it may be an inducible promoter (i.e., a promoter whose state, active/“ON” or inactive/“OFF”, is controlled by an external stimulus, e.g., the presence of a particular temperature, compound, or protein.), it may be a spatially restricted promoter (i.e., transcriptional control element, enhancer, etc.)(e.g., tissue specific promoter, cell type specific promoter, etc.), and it may be a temporally restricted promoter (i.e., the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process, e.g., hair follicle cycle in mice).
  • a constitutively active promoter i.e., a promoter that is constitutively in an active/“ON” state
  • it may be an inducible promote
  • Suitable promoters can be derived from viruses and can therefore be referred to as viral promoters, or they can be derived from any organism, including prokaryotic or eukaryotic organisms. Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • RNA polymerase e.g., pol I, pol II, pol III
  • Exemplary promoters include, but are not limited to the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6) (Miyagishi et al., Nature Biotechnology 20, 497-500 (2002)), an enhanced U6 promoter (e.g., Xia et al., Nucleic Acids Res. 2003 Sep.
  • LTR mouse mammary tumor virus long terminal repeat
  • Ad MLP adenovirus major late promoter
  • HSV herpes simplex virus
  • CMV cytomegalovirus
  • CMVIE CMV immediate early promoter region
  • RSV rous sarcoma virus
  • H1 promoter H1 promoter
  • U6 promoters can be used to control the expression of non-coding RNA molecules (e.g., a guide RNA, an activator-RNA, a targeting-RNA, etc.) in eukaryotic cells.
  • non-coding RNA molecules e.g., a guide RNA, an activator-RNA, a targeting-RNA, etc.
  • inducible promoters include, but are not limited to T7 RNA polymerase promoter, T3 RNA polymerase promoter, Isopropyl-beta-D-thiogalactopyranoside (IPTG)-regulated promoter, lactose induced promoter, heat shock promoter, Tetracycline-regulated promoter, Steroid-regulated promoter, Metal-regulated promoter, estrogen receptor-regulated promoter, etc.
  • Inducible promoters can therefore be regulated by molecules including, but not limited to, doxycycline; RNA polymerase, e.g., T7 RNA polymerase; an estrogen receptor; an estrogen receptor fusion; etc.
  • the promoter is a spatially restricted promoter (i.e., cell type specific promoter, tissue specific promoter, etc.) such that in a multi-cellular organism, the promoter is active (i.e., “ON”) in a subset of specific cells.
  • spatially restricted promoters may also be referred to as enhancers, transcriptional control elements, control sequences, etc.
  • any convenient spatially restricted promoter may be used and the choice of suitable promoter (e.g., a brain specific promoter, a promoter that drives expression in a subset of neurons, a promoter that drives expression in the germline, a promoter that drives expression in the lungs, a promoter that drives expression in muscles, a promoter that drives expression in islet cells of the pancreas, etc.) will depend on the organism.
  • various spatially restricted promoters are known for plants, flies, worms, mammals, mice, etc.
  • a spatially restricted promoter can be used to regulate the expression of a nucleic acid encoding a subject Cas9 protein in a wide variety of different tissues and cell types, depending on the organism.
  • Some spatially restricted promoters are also temporally restricted such that the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process (e.g., hair follicle cycle in mice).
  • spatially restricted promoters include, but are not limited to, neuron-specific promoters, adipocyte-specific promoters, cardiomyocyte-specific promoters, smooth muscle-specific promoters, photoreceptor-specific promoters, etc.
  • Neuron-specific spatially restricted promoters include, but are not limited to, a neuron-specific enolase (NSE) promoter (see, e.g., EMBL HSENO2, X51956); an aromatic amino acid decarboxylase (AADC) promoter; a neurofilament promoter (see, e.g., GenBank HUMNFL, L04147); a synapsin promoter (see, e.g., GenBank HUMSYNIB, M55301); a thy-1 promoter (see, e.g., Chen et al. (1987) Cell 51:7-19; and Llewellyn, et al. (2010) Nat. Med.
  • NSE neuron-specific enolase
  • AADC aromatic amino acid decarboxylase
  • Adipocyte-specific spatially restricted promoters include, but are not limited to aP2 gene promoter/enhancer, e.g., a region from ⁇ 5.4 kb to +21 bp of a human aP2 gene (see, e.g., Tozzo et al. (1997) Endocrinol. 138:1604; Ross et al. (1990) Proc. Natl. Acad. Sci. USA 87:9590; and Pavjani et al. (2005) Nat. Med. 11:797); a glucose transporter-4 (GLUT4) promoter (see, e.g., Knight et al. (2003) Proc. Natl. Acad. Sci.
  • aP2 gene promoter/enhancer e.g., a region from ⁇ 5.4 kb to +21 bp of a human aP2 gene (see, e.g., Tozzo et al. (1997) Endocrinol. 138:160
  • fatty acid translocase (FAT/CD36) promoter see, e.g., Kuriki et al. (2002) Biol. Pharm. Bull. 25:1476; and Sato et al. (2002) J. Biol. Chem. 277:15703
  • SCD1 stearoyl-CoA desaturase-1
  • SCD1 stearoyl-CoA desaturase-1 promoter
  • leptin promoter see, e.g., Mason et al. (1998) Endocrinol. 139:1013; and Chen et al. (1999) Biochem. Biophys. Res. Comm.
  • adiponectin promoter see, e.g., Kita et al. (2005) Biochem. Biophys. Res. Comm. 331:484; and Chakrabarti (2010) Endocrinol. 151:2408
  • an adipsin promoter see, e.g., Platt et al. (1989) Proc. Natl. Acad. Sci. USA 86:7490
  • a resistin promoter see, e.g., Seo et al. (2003) Molec. Endocrinol. 17:1522); and the like.
  • Cardiomyocyte-specific spatially restricted promoters include, but are not limited to control sequences derived from the following genes: myosin light chain-2, ⁇ -myosin heavy chain, AE3, cardiac troponin C, cardiac actin, and the like.
  • Franz et al. (1997) Cardiovasc. Res. 35:560-566; Robbins et al. (1995) Ann. N.Y. Acad. Sci. 752:492-505; Linn et al. (1995) Circ. Res. 76:584-591; Parmacek et al. (1994) Mol. Cell. Biol. 14:1870-1885; Hunter et al. (1993) Hypertension 22:608-617; and Sartorelli et al. (1992) Proc. Natl. Acad. Sci. USA 89:4047-4051.
  • Smooth muscle-specific spatially restricted promoters include, but are not limited to an SM22 ⁇ promoter (see, e.g., Akyürek et al. (2000) Mol. Med. 6:983; and U.S. Pat. No. 7,169,874); a smoothelin promoter (see, e.g., WO 2001/018048); an ⁇ -smooth muscle actin promoter; and the like.
  • a 0.4 kb region of the SM22 ⁇ promoter, within which lie two CArG elements has been shown to mediate vascular smooth muscle cell-specific expression (see, e.g., Kim, et al. (1997) Mol. Cell. Biol. 17, 2266-2278; Li, et al., (1996) J. Cell Biol. 132, 849-859; and Moessler, et al. (1996) Development 122, 2415-2425).
  • Photoreceptor-specific spatially restricted promoters include, but are not limited to, a rhodopsin promoter; a rhodopsin kinase promoter (Young et al. (2003) Ophthalmol. Vis. Sci. 44:4076); a beta phosphodiesterase gene promoter (Nicoud et al. (2007) J. Gene Med. 9:1015); a retinitis pigmentosa gene promoter (Nicoud et al. (2007) supra); an interphotoreceptor retinoid-binding protein (IRBP) gene enhancer (Nicoud et al. (2007) supra); an IRBP gene promoter (Yokoyama et al. (1992) Exp Eye Res. 55:225); and the like.
  • a rhodopsin promoter a rhodopsin kinase promoter
  • a beta phosphodiesterase gene promoter Necoud et al. (2007) J. Gene
  • DNA regulatory sequences refer to transcriptional and translational control sequences, such as promoters, enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence (e.g., guide RNA) or a coding sequence (e.g., a Cas9 protein) and/or regulate translation of an encoded polypeptide.
  • a non-coding sequence e.g., guide RNA
  • a coding sequence e.g., a Cas9 protein
  • nucleic acid refers to a nucleic acid, polypeptide, cell, or organism that is found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by a human in the laboratory is naturally occurring.
  • chimeric refers to two components that are defined by structures derived from different sources.
  • a chimeric polypeptide e.g., a chimeric Cas9 protein
  • the chimeric polypeptide includes amino acid sequences that are derived from different polypeptides.
  • a chimeric polypeptide may comprise either modified or naturally-occurring polypeptide sequences (e.g., a first amino acid sequence from a modified or unmodified Cas9 protein; and a second amino acid sequence other than the Cas9 protein).
  • chimeric in the context of a polynucleotide encoding a chimeric polypeptide includes nucleotide sequences derived from different coding regions (e.g., a first nucleotide sequence encoding a modified or unmodified Cas9 protein; and a second nucleotide sequence encoding a polypeptide other than a Cas9 protein).
  • chimeric polypeptide refers to a polypeptide which is made by the combination (i.e., “fusion”) of two otherwise separated segments of amino sequence, usually through human intervention.
  • a polypeptide that comprises a chimeric amino acid sequence is a chimeric polypeptide.
  • Chimeric polypeptides can be referred to as “fusion variants” or “fusion proteins.”
  • Heterologous means a nucleotide or polypeptide sequence that is not found in the native nucleic acid or protein, respectively.
  • the RNA-binding domain of a naturally-occurring bacterial Cas9 protein (or a variant thereof) may be fused to a heterologous polypeptide sequence (i.e. a polypeptide sequence from a protein other than Cas9 or a polypeptide sequence from another organism).
  • the heterologous polypeptide sequence (also referred to as a “fusion partner”) may exhibit an activity (e.g., enzymatic activity) that will also be exhibited by the chimeric Cas9 protein (e.g., methyltransferase activity, acetyltransferase activity, kinase activity, ubiquitinating activity, etc.).
  • a heterologous nucleic acid sequence may be linked to a naturally-occurring nucleic acid sequence (or a variant thereof) (e.g., by genetic engineering) to generate a chimeric nucleotide sequence encoding a chimeric polypeptide.
  • a variant Cas9 site-directed polypeptide may be fused to a heterologous polypeptide (i.e. a polypeptide other than Cas9), which exhibits an activity that will also be exhibited by the fusion variant Cas9 site-directed polypeptide.
  • a heterologous nucleic acid sequence may be linked to a variant Cas9 site-directed polypeptide (e.g., by genetic engineering) to generate a nucleotide sequence encoding a fusion variant Cas9 site-directed polypeptide.
  • Recombinant means that a particular nucleic acid (DNA or RNA) is the product of various combinations of cloning, restriction, polymerase chain reaction (PCR) and/or ligation steps resulting in a construct having a structural coding or non-coding sequence distinguishable from endogenous nucleic acids found in natural systems.
  • DNA sequences encoding polypeptides can be assembled from cDNA fragments or from a series of synthetic oligonucleotides, to provide a synthetic nucleic acid which is capable of being expressed from a recombinant transcriptional unit contained in a cell or in a cell-free transcription and translation system.
  • Genomic DNA comprising the relevant sequences can also be used in the formation of a recombinant gene or transcriptional unit. Sequences of non-translated DNA may be present 5′ or 3′ from the open reading frame, where such sequences do not interfere with manipulation or expression of the coding regions, and may indeed act to modulate production of a desired product by various mechanisms (see “DNA regulatory sequences”, below). Alternatively, DNA sequences encoding RNA (e.g., guide RNA) that is not translated may also be considered recombinant. Thus, e.g., the term “recombinant” nucleic acid refers to one which is not naturally occurring, e.g., is made by the artificial combination of two otherwise separated segments of sequence through human intervention.
  • This artificial combination is often accomplished by either chemical synthesis means, or by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques. Such is usually done to replace a codon with a codon encoding the same amino acid, a conservative amino acid, or a non-conservative amino acid. Alternatively, it is performed to join together nucleic acid segments of desired functions to generate a desired combination of functions. This artificial combination is often accomplished by either chemical synthesis means, or by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques.
  • a recombinant polynucleotide encodes a polypeptide
  • the sequence of the encoded polypeptide can be naturally occurring (“wild type”) or can be a variant (e.g., a mutant) of the naturally occurring sequence.
  • the term “recombinant” polypeptide does not necessarily refer to a polypeptide whose sequence does not naturally occur.
  • a “recombinant” polypeptide is encoded by a recombinant DNA sequence, but the sequence of the polypeptide can be naturally occurring (“wild type”) or non-naturally occurring (e.g., a variant, a mutant, etc.).
  • a “recombinant” polypeptide is the result of human intervention, but may be a naturally occurring amino acid sequence.
  • a “vector” or “expression vector” is a replicon, such as plasmid, phage, virus, or cosmid, to which another DNA segment, i.e. an “insert”, may be attached so as to bring about the replication of the attached segment in a cell.
  • An “expression cassette” comprises a DNA coding sequence operably linked to a promoter.
  • “Operably linked” refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. For instance, a promoter is operably linked to a coding sequence if the promoter affects its transcription or expression.
  • recombinant expression vector or “DNA construct” are used interchangeably herein to refer to a DNA molecule comprising a vector and at least one insert.
  • Recombinant expression vectors are usually generated for the purpose of expressing and/or propagating the insert(s), or for the construction of other recombinant nucleotide sequences.
  • the insert(s) may or may not be operably linked to a promoter sequence and may or may not be operably linked to DNA regulatory sequences.
  • a cell has been “genetically modified” or “transformed” or “transfected” by exogenous DNA, e.g. a recombinant expression vector, when such DNA has been introduced inside the cell.
  • exogenous DNA e.g. a recombinant expression vector
  • the presence of the exogenous DNA results in permanent or transient genetic change.
  • the transforming DNA may or may not be integrated (covalently linked) into the genome of the cell.
  • the transforming DNA may be maintained on an episomal element such as a plasmid.
  • a stably transformed cell is one in which the transforming DNA has become integrated into a chromosome so that it is inherited by daughter cells through chromosome replication.
  • a “clone” is a population of cells derived from a single cell or common ancestor by mitosis.
  • a “cell line” is a clone of a primary cell that is capable of stable growth in vitro for many generations.
  • Suitable methods of genetic modification include e.g., viral or bacteriophage infection, transfection, conjugation, protoplast fusion, lipofection, electroporation, calcium phosphate precipitation, polyethyleneimine (PEI)-mediated transfection, DEAE-dextran mediated transfection, liposome-mediated transfection, particle gun technology, calcium phosphate precipitation, direct micro injection, nanoparticle-mediated nucleic acid delivery (see, e.g., Panyam et., al Adv Drug Deliv Rev. 2012 Sep. 13. pii: S0169-409X(12)00283-9. doi: 10.1016/j.addr.2012.09.023), and the like.
  • transformation include e.g., viral or bacteriophage infection, transfection, conjugation, protoplast fusion, lipofection, electroporation, calcium phosphate precipitation, polyethyleneimine (PEI)-mediated transfection, DEAE-dextran mediated transfection, liposome-mediated transfection, particle gun technology
  • a “target DNA” as used herein is a DNA polynucleotide that comprises a “target site” or “target sequence.”
  • target site or “target sequence” or “target protospacer DNA” are used interchangeably herein to refer to a nucleic acid sequence present in a target DNA to which a DNA-targeting segment of a subject guide RNA will bind (see FIG. 5A-5B ), provided sufficient conditions for binding exist.
  • the target site (or target sequence) 5′-GAGCATATC-3′ within a target DNA is targeted by (or is bound by, or hybridizes with, or is complementary to) the RNA sequence 5′-GAUAUGCUC-3′.
  • Suitable DNA/RNA binding conditions include physiological conditions normally present in a cell.
  • RNA/RNA binding conditions e.g., conditions in a cell-free system
  • the strand of the target DNA that is complementary to and hybridizes with the guide RNA is referred to as the “complementary strand”
  • the strand of the target DNA that is complementary to the “complementary strand” (and is therefore not complementary to the guide RNA) is referred to as the “noncomplementary strand” or “non-complementary strand.”
  • cleavage it is meant the breakage of the covalent backbone of a DNA molecule. Cleavage can be initiated by a variety of methods including, but not limited to, enzymatic or chemical hydrolysis of a phosphodiester bond. Both single-stranded cleavage and double-stranded cleavage are possible, and double-stranded cleavage can occur as a result of two distinct single-stranded cleavage events. DNA cleavage can result in the production of either blunt ends or staggered ends.
  • a complex comprising a guide RNA and a Cas9 protein is used for targeted double-stranded DNA cleavage. In certain embodiments, a complex comprising a guide RNA and a Cas9 protein is used for targeted cleavage of a single strand of a double-stranded target DNA.
  • Nuclease and “endonuclease” are used interchangeably herein to mean an enzyme which possesses catalytic activity for DNA cleavage.
  • segment it is meant a segment/section/region of a molecule, e.g., a contiguous stretch of nucleotides of an RNA.
  • a segment can also mean a region/section of a complex such that a segment may comprise regions of more than one molecule.
  • the protein-binding segment (described below) of a guide RNA is one RNA molecule and the protein-binding segment therefore comprises a region of that RNA molecule.
  • the protein-binding segment (described below) of a guide RNA comprises two separate molecules that are hybridized along a region of complementarity.
  • a protein-binding segment of a guide RNA that comprises two separate molecules can comprise (i) base pairs 40-75 of a first RNA molecule that is 100 base pairs in length; and (ii) base pairs 10-25 of a second RNA molecule that is 50 base pairs in length.
  • the definition of “segment,” unless otherwise specifically defined in a particular context, is not limited to a specific number of total base pairs, is not limited to any particular number of base pairs from a given RNA molecule, is not limited to a particular number of separate molecules within a complex, and may include regions of RNA molecules that are of any total length and may or may not include regions with complementarity to other molecules.
  • cleavage domain or “active domain” or “nuclease domain” of a nuclease it is meant the polypeptide sequence or domain within the nuclease which possesses the catalytic activity for DNA cleavage.
  • a cleavage domain can be contained in a single polypeptide chain or cleavage activity can result from the association of two (or more) polypeptides.
  • a single nuclease domain may consist of more than one isolated stretch of amino acids within a given polypeptide.
  • Cas9 polypeptide or “Cas9 protein” or “site-directed modifying polypeptide” or “RNA-binding site-directed polypeptide” or “RNA-binding site-directed modifying polypeptide” or “site-directed polypeptide” it is meant a polypeptide that binds RNA and is targeted to a specific DNA sequence.
  • a Cas9 protein as described herein is targeted to a specific DNA sequence by the RNA (a guide RNA) to which it is bound.
  • the guide RNA comprises a sequence that is complementary to a target sequence within the target DNA, thus targeting the bound Cas9 protein to a specific location within the target DNA (the target sequence).
  • guide RNA it is meant an RNA molecule that binds to a Cas9 protein and targets the Cas9 protein to a specific location within the target DNA (see FIG. 5A-5B ).
  • a subject “guide RNA” (also referred to as a “gRNA” or “DNA-targeting RNA” or “DNA-targeting RNA polynucleotide”) has two segments: a “DNA-targeting segment” and a “protein-binding segment.”
  • the DNA-targeting segment of a guide RNA includes a nucleotide sequence (a “DNA-targeting sequence”) that is complementary to a specific sequence within a target DNA (the complementary strand of a target DNA).
  • the protein-binding segment of a guide RNA interacts with a Cas9 protein.
  • the protein-binding segment of a subject guide RNA comprises two complementary stretches of nucleotides that hybridize to one another to form a double stranded RNA duplex (dsRNA duplex).
  • a subject guide RNA comprises two separate RNA molecules (RNA polynucleotides: an “activator-RNA” and a “targeter-RNA”, see below) and is referred to herein as a “dual guide RNA” or a “double-molecule guide RNA” or a “two-molecule guide RNA.”
  • the subject guide RNA is a single RNA molecule (single RNA polynucleotide) and is referred to herein as a “single guide RNA” or “sgRNA” or “single-molecule guide RNA/”
  • the term ⁇ “guide RNA” or “DNA-targeting RNA” or “gRNA” is inclusive, referring both to dual guide RNAs and to single guide RNAs (i.e., sgRNAs).
  • a subject dual guide RNA comprises a “targeter-RNA” (“crRNA-likeRNA” or “CRISPR RNA” or “crRNA” or “crRNA repeat”) molecule and a corresponding “activator-RNA” (“trans-acting CRISPR RNA” or “tracrRNA-like RNA” or “tracrRNA”) molecule.
  • a crRNA-like molecule comprises both the DNA-targeting segment (single stranded) of the guide RNA and a stretch (“duplex-forming segment”) of nucleotides that contribute to the dsRNA duplex of the protein-binding segment of the guide RNA.
  • a corresponding tracrRNA-like molecule also comprises a stretch of nucleotides (duplex-forming segment) that contribute to the dsRNA duplex of the protein-binding segment of the guide RNA.
  • the duplex-forming segment of a targeter-RNA is complementary to and hybridizes with the duplex-forming segment of an activator-RNA to form the dsRNA duplex of the protein-binding domain of the guide RNA.
  • a targeter-RNA molecule can be said to have a corresponding activator-RNA molecule.
  • the targeter-RNA also provides the targeting segment (the segment that hybridizes with the target DNA) of the guide RNA.
  • a targeter-RNA and an activator-RNA hybridize to form a guide RNA.
  • the exact sequence of a given activator-RNA (e.g., a tracrRNA) or targeter-RNA (e.g., a crRNA) molecule can be characteristic of the species in which the RNA molecules are found (or can be derived from such sequences, i.e., truncated, elongated, etc.).
  • Various crRNAs and tracrRNAs are depicted in corresponding complementary pairs e.g., in U.S.
  • a subject guide RNA can comprise any corresponding activator-RNA/targeter-RNA pair (e.g., a crRNA/tracrRNA pair).
  • activator-RNA is used herein to mean a tracrRNA-like molecule (e.g., of a dual guide RNA).
  • targeter-RNA is used herein to mean a crRNA-like molecule (e.g., of a dual guide RNA).
  • duplex-forming segment is used herein to mean the stretch of nucleotides of an activator-RNA or a targeter-RNA that contributes to the formation of the dsRNA duplex by hybridizing to a stretch of nucleotides of a corresponding activator-RNA or targeter-RNA molecule.
  • an activator-RNA comprises a duplex-forming segment that is complementary to the duplex-forming segment of the corresponding targeter-RNA.
  • an activator-RNA comprises a duplex-forming segment while a targeter-RNA comprises both a duplex-forming segment and the DNA-targeting segment of the guide RNA. Therefore, a subject dual guide RNA can be comprised of any corresponding activator-RNA and targeter-RNA pair.
  • Cas9 targeting complex it is meant a complex having a Cas9 protein bound to (i.e., interacting with) a guide RNA.
  • a guide RNA and a subject Cas9 protein form a Cas9 targeting complex (i.e., bind via non-covalent interactions).
  • the guide RNA provides target specificity to the complex by comprising a nucleotide sequence that is complementary to a sequence of a target DNA.
  • the Cas9 protein of the complex provides the site-specific activity.
  • the Cas9 protein is guided to a target DNA sequence (e.g. a target sequence in a chromosomal nucleic acid; a target sequence in an extrachromosomal nucleic acid, e.g.
  • the Cas9 protein has nuclease activity, site-specific cleavage of the target DNA occurs where the Cas9 targeting complex is localized within the target DNA, i.e., at a specific site (i.e., location) in the target DNA determined by the base-pairing complementarity between the DNA-targeting sequence of the guide RNA and the target
  • a subject nucleic acid e.g., a guide RNA, a nucleic acid encoding a guide RNA; a nucleic acid encoding a Cas9 protein; etc.
  • a modification or sequence that provides for an additional desirable feature (e.g., modified or regulated stability; subcellular targeting; tracking, e.g., a fluorescent label; a binding site for a protein or protein complex; etc.).
  • Non-limiting examples include: a 5′ cap (e.g., a 7-methylguanylate cap (m7G)); a 3′ polyadenylated tail (i.e., a 3′ poly(A) tail); a riboswitch sequence (e.g., to allow for regulated stability and/or regulated accessibility by proteins and/or protein complexes); a stability control sequence; a sequence that forms a dsRNA duplex (i.e., a hairpin)); a modification or sequence that targets the RNA to a subcellular location (e.g., nucleus, mitochondria, chloroplasts, and the like); a modification or sequence that provides for tracking (e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.); a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA
  • a subject nucleic acid (e.g., a guide RNA, a nucleic acid encoding a guide RNA; a nucleic acid encoding a Cas9 protein; etc.) comprises an additional segment at either the 5′ or 3′ end that provides for any of the features described above.
  • a suitable third segment can comprise a 5′ cap (e.g., a 7-methylguanylate cap (m7G)); a 3′ polyadenylated tail (i.e., a 3′ poly(A) tail); a riboswitch sequence (e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes); a stability control sequence; a sequence that forms a dsRNA duplex (i.e., a hairpin)); a sequence that targets the RNA to a subcellular location (e.g., nucleus, mitochondria, chloroplasts, and the like); a modification or sequence that provides for tracking (e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.); a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA
  • host cell or “target cell” are used herein to denote an in vivo or in vitro eukaryotic cell (a cell from a unicellular or multicellular organism, e.g., a cell line) which can be, or has been, used as a recipient for a subject Cas9 targeting complex.
  • These terms include the progeny of the original cell which has been targeted (e.g., transfected by nucleic acid encoding a guide RNA). It is understood that the progeny of a single cell may not necessarily be completely identical in morphology or in genomic or total DNA complement as the original parent, due to natural, accidental, or deliberate mutation.
  • a target cell can be any eukaryotic cell having DNA that can be targeted by a Cas9 targeting complex (e.g., a eukaryotic single-cell organism, a somatic cell, a germ cell, a stem cell, a plant cell, an algal cell, an animal cell, in invertebrate cell, a vertebrate cell, a fish cell, a frog cell, a bird cell, a mammalian cell, a pig cell, a cow cell, a goat cell, a sheep cell, a rodent cell, a rat cell, a mouse cell, a non-human primate cell, a human cell, etc.).
  • a Cas9 targeting complex e.g., a eukaryotic single-cell organism, a somatic cell, a germ cell, a stem cell, a plant cell, an algal cell, an animal cell, in invertebrate cell, a vertebrate cell, a fish cell, a frog cell,
  • a “genetically modified host cell” is a host cell into which has been introduced a heterologous nucleic acid, e.g., an expression vector.
  • a subject eukaryotic host cell is a genetically modified eukaryotic host cell (e.g., a mammalian germ cell, a human cell, etc.), by virtue of introduction into the cell of an exogenous nucleic acid (e.g.
  • RNA nucleic acid encoding a guide RNA
  • targeter-RNA nucleic acid encoding a targeter-RNA
  • activator-RNA a nucleic acid encoding an activator-RNA
  • Cas9 protein a nucleic acid encoding a Cas9 protein
  • stem cell is used herein to refer to a cell (e.g., plant stem cell, vertebrate stem cell) that has the ability both to self-renew and to generate a differentiated cell type (see Morrison et al. (1997) Cell 88:287-298).
  • the adjective “differentiated”, or “differentiating” is a relative term.
  • a “differentiated cell” is a cell that has progressed further down the developmental pathway than the cell it is being compared with.
  • pluripotent stem cells can differentiate into lineage-restricted progenitor cells (e.g., mesodermal stem cells), which in turn can differentiate into cells that are further restricted (e.g., neuron progenitors), which can differentiate into end-stage cells (i.e., terminally differentiated cells, e.g., neurons, cardiomyocytes, etc.), which play a characteristic role in a certain tissue type, and may or may not retain the capacity to proliferate further.
  • progenitor cells e.g., mesodermal stem cells
  • end-stage cells i.e., terminally differentiated cells, e.g., neurons, cardiomyocytes, etc.
  • Stem cells may be characterized by both the presence of specific markers (e.g., proteins, RNAs, etc.) and the absence of specific markers.
  • Stem cells may also be identified by functional assays both in vitro and in vivo, particularly assays relating to the ability of stem cells to give rise to multiple differentiated
  • PSCs pluripotent stem cells
  • Pluripotent stem cell or “PSC” is used herein to mean a stem cell capable of producing all cell types of the organism. Therefore, a PSC can give rise to cells of all germ layers of the organism (e.g., the endoderm, mesoderm, and ectoderm of a vertebrate). Pluripotent cells are capable of forming teratomas and of contributing to ectoderm, mesoderm, or endoderm tissues in a living organism. Pluripotent stem cells of plants are capable of giving rise to all cell types of the plant (e.g., cells of the root, stem, leaves, etc.).
  • PSCs of animals can be derived in a number of different ways.
  • embryonic stem cells ESCs
  • iPSCs induced pluripotent stem cells
  • somatic cells Takahashi et. al, Cell. 2007 Nov. 30; 131(5):861-72; Takahashi et. al, Nat Protoc. 2007; 2(12):3081-9; Yu et. al, Science. 2007 Dec. 21; 318(5858):1917-20. Epub 2007 Nov. 20).
  • PSC refers to pluripotent stem cells regardless of their derivation
  • the term PSC encompasses the terms ESC and iPSC, as well as the term embryonic germ stem cells (EGSC), which are another example of a PSC.
  • ESC and iPSC as well as the term embryonic germ stem cells (EGSC), which are another example of a PSC.
  • EGSC embryonic germ stem cells
  • PSCs may be in the form of an established cell line, they may be obtained directly from primary embryonic tissue, or they may be derived from a somatic cell. PSCs can be target cells of the methods described herein.
  • ESC embryonic stem cell
  • ESC lines are listed in the NIH Human Embryonic Stem Cell Registry, e.g.
  • Stem cells of interest also include embryonic stem cells from other primates, such as Rhesus stem cells and marmoset stem cells.
  • the stem cells may be obtained from any mammalian species, e.g.
  • ESCs typically grow as flat colonies with large nucleo-cytoplasmic ratios, defined borders and prominent nucleoli.
  • ESCs express SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, and Alkaline Phosphatase, but not SSEA-1.
  • Examples of methods of generating and characterizing ESCs may be found in, for example, U.S. Pat. Nos. 7,029,913, 5,843,780, and 6,200,806, the disclosures of which are incorporated herein by reference. Methods for proliferating hESCs in the undifferentiated form are described in WO 99/20741, WO 01/51616, and WO 03/020920.
  • EGSC embryonic germ stem cell
  • EG cell a PSC that is derived from germ cells and/or germ cell progenitors, e.g. primordial germ cells, i.e. those that would become sperm and eggs.
  • Embryonic germ cells EG cells
  • Examples of methods of generating and characterizing EG cells may be found in, for example, U.S. Pat. No. 7,153,684; Matsui, Y., et al., (1992) Cell 70:841; Shamblott, M., et al. (2001) Proc. Natl. Acad. Sci.
  • iPSC induced pluripotent stem cell
  • iPSCs can be derived from multiple different cell types, including terminally differentiated cells. iPSCs have an ES cell-like morphology, growing as flat colonies with large nucleo-cytoplasmic ratios, defined borders and prominent nuclei.
  • iPSCs express one or more key pluripotency markers known by one of ordinary skill in the art, including but not limited to Alkaline Phosphatase, SSEA3, SSEA4, Sox2, Oct3/4, Nanog, TRA160, TRA181, TDGF 1, Dnmt3b, FoxD3, GDF3, Cyp26a1, TERT, and zfp42.
  • Examples of methods of generating and characterizing iPSCs may be found in, for example, U.S. Patent Publication Nos. US20090047263, US20090068742, US20090191159, US20090227032, US20090246875, and US20090304646, the disclosures of which are incorporated herein by reference.
  • somatic cells are provided with reprogramming factors (e.g. Oct4, SOX2, KLF4, MYC, Nanog, Lin28, etc.) known in the art to reprogram the somatic cells to become pluripotent stem cells.
  • reprogramming factors e.g. Oct4, SOX2, KLF4, MYC, Nanog, Lin28, etc.
  • somatic cell it is meant any cell in an organism that, in the absence of experimental manipulation, does not ordinarily give rise to all types of cells in an organism.
  • somatic cells are cells that have differentiated sufficiently that they will not naturally generate cells of all three germ layers of the body, i.e. ectoderm, mesoderm and endoderm.
  • somatic cells would include both neurons and neural progenitors, the latter of which may be able to naturally give rise to all or some cell types of the central nervous system but cannot give rise to cells of the mesoderm or endoderm lineages.
  • mitotic cell it is meant a cell undergoing mitosis.
  • Mitosis is the process by which a eukaryotic cell separates the chromosomes in its nucleus into two identical sets in two separate nuclei. It is generally followed immediately by cytokinesis, which divides the nuclei, cytoplasm, organelles and cell membrane into two cells containing roughly equal shares of these cellular components.
  • post-mitotic cell it is meant a cell that has exited from mitosis, i.e., it is “quiescent”, i.e. it is no longer undergoing divisions. This quiescent state may be temporary, i.e. reversible, or it may be permanent.
  • meiotic cell it is meant a cell that is undergoing meiosis.
  • Meiosis is the process by which a cell divides its nuclear material for the purpose of producing gametes or spores. Unlike mitosis, in meiosis, the chromosomes undergo a recombination step which shuffles genetic material between chromosomes. Additionally, the outcome of meiosis is four (genetically unique) haploid cells, as compared with the two (genetically identical) diploid cells produced from mitosis.
  • HDR homology-directed repair
  • Homology-directed repair may result in an alteration of the sequence of the target molecule (e.g., insertion, deletion, mutation), if the donor polynucleotide differs from the target molecule and part or all of the sequence of the donor polynucleotide is incorporated into the target DNA.
  • the donor polynucleotide, a portion of the donor polynucleotide, a copy of the donor polynucleotide, or a portion of a copy of the donor polynucleotide integrates into the target DNA.
  • non-homologous end joining it is meant the repair of double-strand breaks in DNA by direct ligation of the break ends to one another without the need for a homologous template (in contrast to homology-directed repair, which requires a homologous sequence to guide repair). NHEJ often results in the loss (deletion) of nucleotide sequence near the site of the double-strand break.
  • treatment generally mean obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease or symptom in a mammal, and includes: (a) preventing the disease or symptom from occurring in a subject which may be predisposed to acquiring the disease or symptom but has not yet been diagnosed as having it; (b) inhibiting the disease or symptom, i.e., arresting its development; or (c) relieving the disease, i.e., causing regression of the disease.
  • the therapeutic agent may be administered before, during or after the onset of disease or injury.
  • the treatment of ongoing disease, where the treatment stabilizes or reduces the undesirable clinical symptoms of the patient, is of particular interest. Such treatment is desirably performed prior to complete loss of function in the affected tissues.
  • the subject therapy will desirably be administered during the symptomatic stage of the disease, and in some cases after the symptomatic stage of the disease.
  • the terms “individual,” “subject,” “host,” and “patient,” are used interchangeably herein and refer to any mammalian subject for whom diagnosis, treatment, or therapy is desired, particularly humans.
  • the present disclosure provides methods and compositions for site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell.
  • the present disclosure provides methods of binding a target DNA in a eukaryotic cell.
  • the present disclosure provides methods of site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell.
  • the present disclosure provides methods of binding a target DNA in a eukaryotic cell.
  • Subject methods of site-specific modification and/or subject methods of binding can include a step of enriching a cell population for cells that are in a desired phase(s) of the cell cycle (e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.); and a step of contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex).
  • a desired phase(s) of the cell cycle e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.
  • the subject methods include contacting the target DNA in the target cell(s) with: (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein.
  • a step of enriching can include, for example, a cell separation method and/or a cell synchronization method.
  • a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at a desired phase in the cell cycle (e.g., the S-phase, the M-phase, the G0 phase, the G1 phase, the G2 phase, G1/S, G2/M, etc.); and a step of contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex).
  • blocking a cell at a phase in the cell cycle includes contacting the cell with a cell cycle blocking agent.
  • a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at the S-phase. In some embodiments, a subject method of site-specific modification and/or a subject method of target nucleic acid binding includes a step of blocking a target eukaryotic cell at the M-phase. In some embodiments, a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at the G0 phase. In some embodiments, a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at the G1 phase.
  • a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at the G2 phase. In some embodiments, a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at G1/S. In some embodiments, a subject method of site-specific modification and/or a subject method of binding includes a step of blocking a target cell at G2/M.
  • a subject method of site-specific modification and/or a subject method of target nucleic acid binding comprising: a) blocking a target cell at a desired phase in the cell cycle; and b) contacting target DNA in the target cell with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein increases the efficiency of HDR and/or NHEJ by at least 10%, at least 25%, at least 50%, at least 75%, at least 2-fold, at least 5-fold, at least 10-fold, or more than 10-fold, compared to the efficiency of site-specific modification or target nucleic acid binding carried out in the absence of a cell cycle blocking or enriching step.
  • the “efficiency” of non-homologous end joining (NHEJ) and/or homology directed repair (HDR) can be calculated by any convenient method. For example, in some cases, efficiency can be expressed in terms of percentage of successful HDR.
  • a restriction digest assay e.g., using a restriction enzyme such as HindIII
  • a restriction enzyme can be used to generate cleavage products and the ratio of products to substrate can be used to calculate the percentage.
  • a restriction enzyme can be used that directly cleaves DNA containing a newly integrated restriction sequence as the result of successful HDR. More cleaved substrate indicates a greater percent HDR (a greater efficiency of HDR).
  • a fraction (percentage) of HDR can be calculated using the following equation [(cleavage products)/(substrate plus cleavage products)] (e.g., b+c/a+b+c), where “a” is the band intensity of DNA substrate and “b” and “c” are the cleavage products.
  • efficiency can be expressed in terms of percentage of successful NHEJ.
  • a T7 endonuclease I assay can be used to generate cleavage products and the ratio of products to substrate can be used to calculate the percentage NHEJ.
  • T7 endonuclease I cleaves mismatched heteroduplex DNA which arises from hybridization of wild-type and mutant DNA strands (NHEJ generates small random insertions or deletions (indels) at the site of the original break). More cleavage indicates a greater percent NHEJ (a greater efficiency of NHEJ).
  • a fraction (percentage) of NHEJ can be calculated using the following equation: (1 ⁇ (1 ⁇ (b+c/a+b+c)) 1/2 ) ⁇ 100, where “a” is the band intensity of DNA substrate and “b” and “c” are the cleavage products (see Example 1 and see Ran et. al., Cell. 2013 Sep. 12; 154(6):1380-9).
  • This formula is used (instead of the formula used for HDR, see above) because upon re-annealing, one duplex of mutant DNA can produce two duplexes of mutant:wild-type hybrid, doubling the actual NHEJ frequency.
  • a subject method of site-specific modification and/or a subject method of target nucleic acid binding comprising: a) contacting a target cell with a cell cycle blocking agent that blocks the target cell at a desired phase in the cell cycle; and b) contacting target DNA in the target cell with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein increases the efficiency of HDR and/or NHEJ by at least 10%, at least 25%, at least 50%, at least 75%, at least 2-fold, at least 5-fold, at least 10-fold, or more than 10-fold, compared to the efficiency of site-specific modification or target nucleic acid binding carried out in the absence of the cell cycle blocking agent.
  • a subject method of site-specific modification and/or a subject method of target nucleic acid binding comprises: a) contacting a target cell with a cell cycle blocking agent that blocks the target cell at a desired phase in the cell cycle; and b) contacting target DNA in the target cell with a Cas9 protein/sgRNA complex.
  • a subject method of site-specific modification and/or a subject method of target nucleic acid binding comprising: a) contacting a target cell with a cell cycle blocking agent that blocks the target cell at a desired phase in the cell cycle; and b) contacting target DNA in the target cell with a Cas9 protein/sgRNA complex increases the efficiency of HDR and/or NHEJ by at least 10%, at least 25%, at least 50%, at least 75%, at least 2-fold, at least 5-fold, at least 10-fold, or more than 10-fold, compared to the efficiency of site-specific modification or target nucleic acid binding carried out in the absence of the cell cycle blocking agent.
  • the present disclosure provides a method of site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell, the method comprising: (a) blocking the cell at a desired phase in the cell cycle; and (b) contacting the target DNA in the cell with: (i) a Cas9 protein, and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding domain that interacts with the Cas9 protein.
  • a subject method comprises contacting target DNA in the eukaryotic cell with a Cas9 protein/sgRNA complex.
  • a subject method of site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell comprises: (a) blocking the cell at S phase; and (b) contacting the target DNA in the cell with: (i) a Cas9 protein, and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding domain that interacts with the Cas9 protein.
  • a subject method of site-specific modification of a target DNA, or a protein associated with a target DNA, in a eukaryotic cell comprises: (a) blocking the cell at M phase; and (b) contacting the target DNA in the cell with: (i) a Cas9 protein, and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding domain that interacts with the Cas9 protein.
  • a subject method of site-specific modification and/or a subject method of target nucleic acid binding comprises: a) contacting a target cell with a cell cycle blocking agent that blocks the target cell at a desired phase in the cell cycle; and b) contacting target DNA in the target cell with a Cas9 protein/guide RNA complex (e.g., Cas9 protein/sgRNA; Cas9 protein/dual-guide RNA, etc.).
  • a Cas9 protein/guide RNA complex e.g., Cas9 protein/sgRNA; Cas9 protein/dual-guide RNA, etc.
  • a cell can be contacted with a Cas9 protein/guide RNA complex having a molar ration, of Cas9 protein to guide RNA, in a range of from 5:1 to 1:5 (e.g., from 4:1 to 1:4, from 3:1 to 1:3, from 2:1 to 1:2, from 1.5:1 to 1:1.5, from 1:1 to 1:1.5, or from 1:1.1 to 1:1.3).
  • a cell can be contacted with a Cas9 protein/guide RNA complex having a molar ration, of Cas9 protein to guide RNA, of 1:1, 1:1.1, 1:1.2, 1:1.3, or 1:1.4.
  • a cell can be contacted with a Cas9 protein/guide RNA complex having a molar ration, of Cas9 protein to guide RNA, of 1:1.2.
  • the amount of Cas9 used to generate the targeting complex is in a range of from 1 pmol to 500 pmol (e.g., from 10 pmol to 400 pmol, from 10 pmol to 300 pmol, from 10 pmol to 250 pmol, from 10 pmol to 200 pmol, from 20 pmol to 200 pmol, from 20 pmol to 150 pmol, from 30 pmol to 100 pmol, from 50 pmol to 100 pmol, or from 60 pmol to 100 pmol) (e.g., see materials and methods section of Example 1 below).
  • 1 pmol to 500 pmol e.g., from 10 pmol to 400 pmol, from 10 pmol to 300 pmol, from 10 pmol to 250 pmol, from 10 pmol to 200 pmol, from 20 pmol to 200 pmol, from 20 pmol to 150 pmol, from 30 pmol to 100 pmol, from 50 pmol to 100 pmol
  • a cell can be contacted with an amount of Cas9 protein/guide RNA in a range of from 1 pmol to 500 pmol (e.g., from 10 pmol to 400 pmol, from 10 pmol to 300 pmol, from 10 pmol to 250 pmol, from 10 pmol to 200 pmol, from 20 pmol to 200 pmol, from 20 pmol to 150 pmol, from 30 pmol to 100 pmol, from 50 pmol to 100 pmol, or from 60 pmol to 100 pmol).
  • 1 pmol to 500 pmol e.g., from 10 pmol to 400 pmol, from 10 pmol to 300 pmol, from 10 pmol to 250 pmol, from 10 pmol to 200 pmol, from 20 pmol to 200 pmol, from 20 pmol to 150 pmol, from 30 pmol to 100 pmol, from 50 pmol to 100 pmol, or from 60 pmol to 100
  • subject methods include (i) the step of enriching a cell population for cells that are in a desired phase(s) of the cell cycle, and/or (ii) the step of blocking a cell at a desired phase in the cell cycle.
  • the cell cycle is the series of events that take place in a cell leading to its division and duplication (replication) that produces two daughter cells.
  • Two major phases of the cell cycle are the S phase (DNA synthesis phase), in which DNA duplication occurs, and the M phase (mitosis), in which the chromosomes segregation and cell division occurs.
  • the eukaryotic cell cycle is traditionally divided into four sequential phases: G1, S, G2, and M. G1, S, and G2 together can collectively be referred to as “interphase”.
  • G1 a specialized resting state known as G0 (G zero), in which they can remain for days, weeks, or even years before resuming proliferation.
  • G0 a specialized resting state
  • the period of transition from one state to another can be referred to using a hyphen, for example, G1/S, G2/M, etc.
  • various checkpoints exist throughout the cell cycle at which a cell can monitor conditions to determine whether cell cycle progression should occur.
  • the G2/M DNA damage checkpoint serves to prevent cells from entering mitosis (M-phase) with genomic DNA damage.
  • a step of enriching a population of eukaryotic cells for cells in a desired phase of the cell cycle can be performed using any convenient method (e.g., a cell separation method and/or a cell synchronization method).
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the G0 phase of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the G0 phase of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to a target
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the G1 phase of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the G1 phase of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to a target
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the G2 phase of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the G2 phase of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to a target
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the S phase of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the S phase of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the M phase of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the M phase of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the G1/S transition of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the G1/S transition of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to
  • a subject method includes a step of enriching a population of eukaryotic cells for cells in the G2/M transition of the cell cycle.
  • a subject method includes: (a) enriching a population of eukaryotic cells for cells in the G2/M transition of the cell cycle; and (b) contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex)(e.g., contacting the target DNA with (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex
  • a guide RNA comprising: a targeting sequence that hybridizes to
  • enriching is meant increasing the fraction of desired cells in the resulting cell population. For example, in some cases, enriching includes selecting desirable cells (e.g., cells that are in the desired phase of the cell cycle) away from undesirable cells (e.g., cells that are not in the desired phase of the cell cycle), which can result in a smaller population of cells, but a greater fraction (i.e., higher percentage) of the cells of the resulting cell population will be desirable cells (e.g., cells that are in the desired phase of the cell cycle).
  • desirable cells e.g., cells that are in the desired phase of the cell cycle
  • undesirable cells e.g., cells that are not in the desired phase of the cell cycle
  • enriching includes converting undesirable cells (e.g., cells that are not in the desired phase of the cell cycle) into desirable cells (e.g., cells that are in the desired phase of the cell cycle), which can result in a similar size population of cells as the starting population, but a greater fraction of those cells will be desirable cells (e.g., cells that are in the desired phase of the cell cycle).
  • Cell synchronization methods (described below) can be an example of this type of enrichment.
  • enrichment can both change the overall size of the resulting cell population (compared to the size of the starting population) and increase the fraction of desirable cells. For example, multiple methods/techniques can be combined (e.g., to improve enrichment, to enrich for cells a more than one desired phase of the cell cycle, etc.).
  • enriching includes a cell separation method.
  • Any convenient cell separation method can be used to enrich for cells that are at various phases of the cell cycle.
  • Suitable cell separation techniques for enrichment of cells at particular phases of the cell cycle include, but are not limited to: (i) mitotic shake-off (M-phase; mechanical separation on the basis of cell adhesion properties, e.g., adherent cells in the mitotic phase detach from the surface upon gentle shaking, tapping, or rinsing); (ii) Countercurrent centrifugal elutriation (CCE) (G1, S, G2/M, and intermediate states; physical separation on the basis of cell size and density); and (iii) flow cytometry and cell sorting (e.g., G0, G1, S, G2/M; physical separation based on specific intracellular, e.g., DNA, content) and cell surface and/or size properties).
  • M-phase mitotic shake-off
  • CCE Countercurrent centrifugal elutriation
  • Mitotic shake-off generally includes dislodgment of low adhesive, mitotic cells by agitation (see for example, Beyrouthy et. al., PLoS ONE 3, e3943 (2008); Schorl, C. & Sedivy, Methods 41, 143-150 (2007)).
  • CCE generally includes the separation of cells according to their sedimentation velocity in a gravitational field where the liquid containing the cells is made to flow against the centrifugal force with the sedimentation rate of cells being proportional to their size (see for example, Grosse et. al., Prep Biochem Biotechnol. 2012; 42(3):217-33; Banfalvi et. al., Nat. Protoc. 3, 663-673 (2008)).
  • Flow cytometry methods generally include the characterization of cells according to antibody and/or ligand and/or dye-mediated fluorescence and scattered light in a hydrodynamically focused stream of liquid with subsequent electrostatic, mechanical or fluidic switching sorting (see for example, Coán et. al., Biochem. Pharmacol. 72, 1396-1404 (2006); Juan et. al., Cytometry 49, 170-175 (2002)).
  • electrostatic, mechanical or fluidic switching sorting see for example, Cottle et. al., Biochem. Pharmacol. 72, 1396-1404 (2006); Juan et. al., Cytometry 49, 170-175 (2002).
  • Rosner et al., Nat Protoc. 2013 March; 8(3):602-26 For more information related to cell separation techniques, refer to, for example, Rosner et al., Nat Protoc. 2013 March; 8(3):602-26.
  • enriching includes a cell synchronization method (i.e., synchronizing the cells of a cell population).
  • Cell synchronization is a process by which cells at different stages of the cell cycle within a cell population (i.e., a population of cells in which various individual cells are in different phases of the cycle) are brought into the same phase.
  • Any convenient cell synchronization method can be used in the subject methods to enrich for cells that are at a desired phase(s) of the cell cycle.
  • cell synchronization can be achieved by blocking cells at a desired phase in the cell cycle, which allows the other cells to cycle until they reach the blocked phase.
  • suitable methods of cell synchronization include, but are not limited to: (i) inhibition of DNA replication, DNA synthesis, and/or mitotic spindle formation (e.g., sometimes referred to herein as contacting a cell with a cell cycle blocking composition); (ii) mitogen or growth factor withdrawal (G0, G1, G0/G1; growth restriction-induced quiescence via, e.g., serum starvation and/or amino acid starvation); and (iii) density arrest (G1; cell-cell contact-induced activation of specific transcriptional programs) (see for example, Rosner et al., Nat Protoc. 2013 March; 8(3):602-26 (e.g., see Table 1 of Rosner et al.), which is hereby incorporated by reference in its entirety, and see references cited therein).
  • a cell is blocked at a desired phase of the cell cycle (e.g., by contacting the cell with a cycle blocking composition).
  • cells of a cell population are synchronized (e.g., by contacting the cells with a cell cycle blocking composition).
  • a cell cycle blocking composition can include one or more cell cycle blocking agents.
  • the term “cell cycle blocking agent” is used herein to refer to an agent that blocks (e.g., reversibly blocks (pauses), irreversibly blocks) a cell at a particular point in the cell cycle such that the cell cannot proceed further.
  • Suitable cell cycle blocking agents include reversible cell cycle blocking agents. Reversible cell cycle blocking agents do not render the cell permanently blocked.
  • Cell cycle blocking agents are sometimes referred to in the art as cell synchronization agents because when such agents contact a cell population (e.g., a population having cells that are at different stages of the cell cycle), the cells of the population become blocked at the same phase of the cell cycle, thus synchronizing the population of cells relative to that particular phase of the cell cycle.
  • the cell cycle blocking agent used is reversible, the cells can then be “released” from cell cycle block.
  • Suitable cell cycle blocking agents include, but are not limited to: nocodazole (G2, M, G2/M; inhibition of microtubule polymerization), colchicine (G2, M, G2/M; inhibition of microtubule polymerization); demecolcine (colcemid) (G2, M, G2/M; inhibition of microtubule polymerization); hydroxyurea (G1, S, G1/S; inhibition of ribonucleotide reductase); aphidicolin (G1, S, G1/S; inhibition of DNA polymerase- ⁇ and DNA polymerase- ⁇ ); lovastatin (G1; inhibition of HMG-CoA reductase/cholesterol synthesis and the proteasome); mimosine (G1, S, G1/S; inhibition of thymidine, nucleotide biosynthesis, inhibition of Ctf4/chromatin binding); thymidine (G1, S, G1/S; excess thymidine-induced feedback inhibition of DNA replication); latrunculin A (
  • Suitable cell cycle blocking agents can include any agent that has the same or similar function as the agents above (e.g., an agent that inhibits microtubule polymerization, an agent that inhibits ribonucleotide reductase, an agent that inhibits DNA polymerase- ⁇ and/or DNA polymerase- ⁇ , an agent that inhibits HMG-CoA reductase and/or cholesterol synthesis, an agent that inhibits nucleotide biosynthesis, an agent that inhibits DNA replication, i.e., inhibit DNA synthesis, an agent that inhibits initiation of DNA replication, an agent that inhibits deoxycytosine synthesis, an agent that induces excess thymidine-induced feedback inhibition of DNA replication, and agent that disrupts interpolar microtubule stability, an agent that inhibits actin polymerization, and the like).
  • an agent that inhibits microtubule polymerization e.g., an agent that inhibits ribonucleotide reductase, an agent that inhibits DNA polymerase-
  • Suitable agents that block G1 can include: staurosporine, dimethyl sulfoxide (DMSO), glycocorticosteroids, and/or mevalonate synthesis inhibitors.
  • Suitable agents that block G2 phase can include CDK1 inhibitors e.g., RO-3306.
  • Suitable agents that block M can include cytochalasin D.
  • suitable cell cycle blocking agents include: cobtorin; dinitroaniline; benefin (benluralin); butralin; dinitramine; ethalfluralin; oryzalin; pendimethalin; trifluralin; amiprophos-methyl; butamiphos dithiopyr; thiazopyr propyzamider-pronamide-tebutam DCPA (chlorthal-dimethyl); anisomycin; alpha amanitin; jasmonic acid; abscisic acid; menadione; cryptogeine; hydrogen peroxide; sodium permanganate; indomethacin; epoxomycin; lactacystein; icrf 193; olomoucine; roscovitine; bohemine; K252a; okadaic acid; endothal; caffeine; MG132; cycline dependent kinase inhibitors; and the like.
  • the subject methods include a step of contacting the target DNA with a Cas9 targeting complex (e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex).
  • a Cas9 targeting complex e.g., via introducing into the target eukaryotic cell(s) at least one component of a Cas9 targeting complex.
  • the subject methods can include contacting the target DNA in the target cell(s) with: (i) a Cas9 protein; and (ii) a guide RNA comprising: a targeting sequence that hybridizes to a target sequence of the target DNA, and a protein-binding segment that interacts with the Cas9 protein.
  • a subject guide RNA that directs the activities of an associated polypeptide (e.g., a Cas9 protein) to a specific target sequence within a target DNA.
  • a subject guide RNA comprises: a first segment (also referred to herein as a “nucleic acid targeting segment”, or simply a “targeting segment”); and a second segment (also referred to herein as a “protein-binding segment”).
  • the first segment of a subject guide RNA comprises a nucleotide sequence that can be complementary to a sequence (a target site) in a target DNA.
  • the targeting segment of a subject guide RNA can interact with a target DNA in a sequence-specific manner via hybridization (i.e., base pairing).
  • the nucleotide sequence of the targeting segment may vary and can determine the location within the target DNA that the guide RNA and the target DNA will interact.
  • the targeting segment of a subject guide RNA can be modified (e.g., by genetic engineering) to hybridize to any desired sequence (target site) within a target DNA.
  • the targeting segment can have a length of from about 12 nucleotides to about 100 nucleotides.
  • the targeting segment can have a length of from about 12 nucleotides (nt) to about 80 nt, from about 12 nt to about 50 nt, from about 12 nt to about 40 nt, from about 12 nt to about 30 nt, from about 12 nt to about 25 nt, from about 12 nt to about 20 nt, or from about 12 nt to about 19 nt.
  • the targeting segment can have a length of from about 19 nt to about 20 nt, from about 19 nt to about 25 nt, from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from about 19 nt to about 40 nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt, from about 19 nt to about 60 nt, from about 19 nt to about 70 nt, from about 19 nt to about 80 nt, from about 19 nt to about 90 nt, from about 19 nt to about 100 nt, from about 20 nt to about 25 nt, from about 20 nt to about 30 nt, from about 20 nt to about 35 nt, from about 20 nt to about 40 nt, from about 20 nt to about 45 nt, from about 20 nt to about 50 nt, from about 20 nt to about 60
  • the nucleotide sequence (the targeting sequence) of the targeting segment that is complementary to a nucleotide sequence (target site) of the target DNA can have a length of 12 nt or more.
  • the targeting sequence of the targeting segment that is complementary to a target site of the target DNA can have a length of 12 nt or more, 15 nt or more, 18 nt or more, 19 nt or more, 20 nt or more, 25 nt or more, 30 nt or more, 35 nt or more or 40 nt.
  • the targeting sequence of the targeting segment that is complementary to a target sequence of the target DNA can have a length of from about 12 nucleotides (nt) to about 80 nt, from about 12 nt to about 50 nt, from about 12 nt to about 45 nt, from about 12 nt to about 40 nt, from about 12 nt to about 35 nt, from about 12 nt to about 30 nt, from about 12 nt to about 25 nt, from about 12 nt to about 20 nt, from about 12 nt to about 19 nt, from about 19 nt to about 20 nt, from about 19 nt to about 25 nt, from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from about 19 nt to about 40 nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt, from about 19 nt to about 60 nt,
  • the targeting sequence of the targeting segment that is complementary to a target site of the target DNA is 20 nucleotides in length. In some cases, the targeting sequence of the targeting segment that is complementary to a target site of the target DNA is 19 nucleotides in length.
  • the percent complementarity between the targeting sequence of the targeting segment and the target site of the target DNA can be 60% or more (e.g., 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% or more, or 100%). In some cases, the percent complementarity between the targeting sequence of the targeting segment and the target site of the target DNA is 100% over the seven contiguous 5′-most nucleotides of the target site of the target DNA. In some cases, the percent complementarity between the targeting sequence of the targeting segment and the target site of the target DNA is 60% or more over about 20 contiguous nucleotides.
  • the percent complementarity between the targeting sequence of the targeting segment and the target site of the target DNA is 100% over the fourteen contiguous 5′-most nucleotides of the target site of the target DNA and as low as 0% or more over the remainder. In such a case, the targeting sequence can be considered to be 14 nucleotides in length. In some cases, the percent complementarity between the targeting sequence of the targeting segment and the target site of the target DNA is 100% over the seven contiguous 5′-most nucleotides of the target site of the target DNA and as low as 0% or more over the remainder. In such a case, the targeting sequence can be considered to be 20 nucleotides in length.
  • the protein-binding segment of a subject guide RNA interacts with a Cas9 protein.
  • the subject guide RNA guides the bound polypeptide to a specific nucleotide sequence within target DNA via the above mentioned targeting segment.
  • the protein-binding segment of a subject guide RNA comprises two stretches of nucleotides that are complementary to one another. The complementary nucleotides of the protein-binding segment hybridize to form a double stranded RNA duplex (dsRNA) (see FIG. 5A-5B ).
  • a subject dual guide RNA comprises two separate nucleic acid molecules.
  • Each of the two molecules of a subject dual guide RNA comprises a stretch of nucleotides that are complementary to one another such that the complementary nucleotides of the two molecules hybridize to form the double stranded RNA duplex of the protein-binding segment ( FIG. 5A ).
  • the duplex-forming segment of the activator-RNA is 60% or more identical to one of the activator-RNA molecules (e.g., tracrRNA molecules) set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • the activator-RNA molecules e.g., tracrRNA molecules set forth in SEQ ID NOs:431-562, or a complement thereof
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 65% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 70% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 75% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 80% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 85% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 90% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 95% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 98% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 99% or more identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the activator-RNA (or the DNA encoding the duplex-forming segment of the activator-RNA) can be 100% identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA is 60% or more identical to one of the targeter-RNA (e.g., crRNA) sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • the targeter-RNA e.g., crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 65% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 70% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 75% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 80% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 85% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 90% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 95% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 98% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 99% or more identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • the duplex-forming segment of the targeter-RNA (or the DNA encoding the duplex-forming segment of the targeter-RNA) can be 100% identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • a dual guide RNA can be designed to allow for controlled (i.e., conditional) binding of a targeter-RNA with an activator-RNA. Because a dual guide RNA is not functional unless both the activator-RNA and the targeter-RNA are bound in a functional complex with Cas9, a dual guide RNA can be inducible (e.g., drug inducible) by rendering the binding between the activator-RNA and the targeter-RNA to be inducible.
  • RNA aptamers can be used to regulate (i.e., control) the binding of the activator-RNA with the targeter-RNA. Accordingly, the activator-RNA and/or the targeter-RNA can include an RNA aptamer sequence.
  • RNA aptamers are known in the art and are generally a synthetic version of a riboswitch.
  • the terms “RNA aptamer” and “riboswitch” are used interchangeably herein to encompass both synthetic and natural nucleic acid sequences that provide for inducible regulation of the structure (and therefore the availability of specific sequences) of the nucleic acid molecule (e.g., RNA, DNA/RNA hybrid, etc.) of which they are part.
  • RNA aptamers usually comprise a sequence that folds into a particular structure (e.g., a hairpin), which specifically binds a particular drug (e.g., a small molecule).
  • Binding of the drug causes a structural change in the folding of the RNA, which changes a feature of the nucleic acid of which the aptamer is a part.
  • an activator-RNA with an aptamer may not be able to bind to the cognate targeter-RNA unless the aptamer is bound by the appropriate drug
  • a targeter-RNA with an aptamer may not be able to bind to the cognate activator-RNA unless the aptamer is bound by the appropriate drug
  • a targeter-RNA and an activator-RNA, each comprising a different aptamer that binds a different drug may not be able to bind to each other unless both drugs are present.
  • a dual guide RNA can be designed to be inducible.
  • aptamers and riboswitches can be found, for example, in: Nakamura et al., Genes Cells. 2012 May; 17(5):344-64; Vavalle et al., Future Cardiol. 2012 May; 8(3):371-82; Citartan et al., Biosens Bioelectron. 2012 Apr. 15; 34(1):1-11; and Liberman et al., Wiley Interdiscip Rev RNA. 2012 May-June; 3(3):369-84; all of which are herein incorporated by reference in their entirety.
  • Non-limiting examples of nucleotide sequences that can be included in a dual guide RNA include either of the sequences set forth in SEQ ID NOs:431-562, or complements thereof pairing with any sequences set forth in SEQ ID NOs:563-679, or complements thereof that can hybridize to form a protein binding segment.
  • a subject single guide RNA comprises two stretches of nucleotides (much like a “targeter-RNA” and an “activator-RNA” of a dual guide RNA) that are complementary to one another, hybridize to form the double stranded RNA duplex (dsRNA duplex) of the protein-binding segment (thus resulting in a stem-loop structure), and are covalently linked by intervening nucleotides (“linkers” or “linker nucleotides”).
  • the targeter-RNA and the activator-RNA can be covalently linked via the 3′ end of the targeter-RNA and the 5′ end of the activator-RNA.
  • targeter-RNA and the activator-RNA can be covalently linked via the 5′ end of the targeter-RNA and the 3′ end of the activator-RNA.
  • the linker of a single guide RNA can have a length of from about 3 nucleotides to about 100 nucleotides.
  • the linker can have a length of from about 3 nucleotides (nt) to about 90 nt, from about 3 nucleotides (nt) to about 80 nt, from about 3 nucleotides (nt) to about 70 nt, from about 3 nucleotides (nt) to about 60 nt, from about 3 nucleotides (nt) to about 50 nt, from about 3 nucleotides (nt) to about 40 nt, from about 3 nucleotides (nt) to about 30 nt, from about 3 nucleotides (nt) to about 20 nt or from about 3 nucleotides (nt) to about 10 nt.
  • the linker can have a length of from about 3 nt to about 5 nt, from about 5 nt to about 10 nt, from about 10 nt to about 15 nt, from about 15 nt to about 20 nt, from about 20 nt to about 25 nt, from about 25 nt to about 30 nt, from about 30 nt to about 35 nt, from about 35 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt.
  • the linker of a single guide RNA is 4 nt.
  • An exemplary single guide RNA comprises two complementary stretches of nucleotides that hybridize to form a dsRNA duplex.
  • one of the two complementary stretches of nucleotides of the single guide RNA (or the DNA encoding the stretch) is 60% or more identical to one of the activator-RNA (e.g., tracrRNA) molecules set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • one of the two complementary stretches of nucleotides of the single guide RNA is 65% or more identical, 70% or more identical, 75% or more identical, 80% or more identical, 85% or more identical, 90% or more identical, 95% or more identical, 98% or more identical, 99% or more identical or 100% identical to one of the tracrRNA sequences set forth in SEQ ID NOs:431-562, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides,
  • one of the two complementary stretches of nucleotides of the single guide RNA is 60% or more identical to one of the targeter-RNA (e.g., crRNA) sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • the targeter-RNA e.g., crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof
  • one of the two complementary stretches of nucleotides of the single guide RNA is 65% or more identical, 70% or more identical, 75% or more identical, 80% or more identical, 85% or more identical, 90% or more identical, 95% or more identical, 98% or more identical, 99% or more identical or 100% identical to one of the crRNA sequences set forth in SEQ ID NOs:563-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or
  • one of the two complementary stretches of nucleotides of the single guide RNA is 60% or more identical to one of the targeter-RNA (e.g., crRNA) sequences or activator-RNA (e.g., tracrRNA) sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • the targeter-RNA e.g., crRNA
  • activator-RNA e.g., tracrRNA
  • one of the two complementary stretches of nucleotides of the single guide RNA can be 65% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 70% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 75% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides)
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 80% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 85% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 90% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 95% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 98% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 99% or more identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • 8 or more contiguous nucleotides e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides.
  • One of the two complementary stretches of nucleotides of the single guide RNA can be 100% identical to one of the sequences set forth in SEQ ID NOs: 431-679, or a complement thereof, over a stretch of 8 or more contiguous nucleotides (e.g., 8 or more contiguous nucleotides, 10 or more contiguous nucleotides, 12 or more contiguous nucleotides, 15 or more contiguous nucleotides, or 20 or more contiguous nucleotides).
  • Appropriate cognate pairs of targeter-RNAs and activator-RNAs can be routinely determined for SEQ ID NOs:431-679 by taking into account the species name and base-pairing (for the dsRNA duplex of the protein-binding domain). See, for example, U.S.
  • patent applications 20140068797, 20140189896, 20140179006, 20140170753, 20140179770, 20140186958, 20140186919, 20140186843; international applications: WO2013176772, WO2013141680, WO2013142578, WO2014065596, WO2014089290, WO2014099744, WO2014099750, WO2014104878, WO2014093718, WO2014093622, WO2014093655, WO2014093701, WO2014093712, WO2014093635, WO2014093595, WO2014093694, and WO2014093661; and U.S. Pat. Nos.
  • activator-RNA sequences 8,697,359, 8,771,945 for non-limiting examples of activator-RNA sequences, targeter-RNA sequences, paired activator-RNA/targeter-RNA sequences, and single guide RNA sequences designed from corresponding activator-RNA/targeter-RNA pairs).
  • the protein-binding segment can have a length of from about 10 nucleotides to about 100 nucleotides.
  • the protein-binding segment can have a length of from about 15 nucleotides (nt) to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt or from about 15 nt to about 25 nt.
  • the dsRNA duplex of the protein-binding segment can have a length from about 6 base pairs (bp) to about 50 bp.
  • the dsRNA duplex of the protein-binding segment can have a length from about 6 bp to about 40 bp, from about 6 bp to about 30 bp, from about 6 bp to about 25 bp, from about 6 bp to about 20 bp, from about 6 bp to about 15 bp, from about 8 bp to about 40 bp, from about 8 bp to about 30 bp, from about 8 bp to about 25 bp, from about 8 bp to about 20 bp or from about 8 bp to about 15 bp.
  • the dsRNA duplex of the protein-binding segment can have a length from about from about 8 bp to about 10 bp, from about 10 bp to about 15 bp, from about 15 bp to about 18 bp, from about 18 bp to about 20 bp, from about 20 bp to about 25 bp, from about 25 bp to about 30 bp, from about 30 bp to about 35 bp, from about 35 bp to about 40 bp, or from about 40 bp to about 50 bp.
  • the dsRNA duplex of the protein-binding segment has a length of 36 base pairs.
  • the percent complementarity between the nucleotide sequences that hybridize to form the dsRNA duplex of the protein-binding segment can be 60% or more.
  • the percent complementarity between the nucleotide sequences that hybridize to form the dsRNA duplex of the protein-binding segment can be 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, or 99% or more.
  • the percent complementarity between the nucleotide sequences that hybridize to form the dsRNA duplex of the protein-binding segment is 100%.
  • Stability Control Sequence e.g., Transcriptional Terminator Segment
  • a guide RNA comprises a stability control sequence.
  • a stability control sequence influences the stability of a nucleic acid (e.g., a guide RNA, a targeter-RNA, an activator-RNA, etc.).
  • a suitable stability control sequence for use with an RNA is a transcriptional terminator segment (i.e., a transcription termination sequence).
  • a transcriptional terminator segment of a subject guide RNA can have a total length of from about 10 nucleotides to about 100 nucleotides, e.g., from about 10 nucleotides (nt) to about 20 nt, from about 20 nt to about 30 nt, from about 30 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt.
  • the transcriptional terminator segment can have a length of from about 15 nucleotides (nt) to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt or from about 15 nt to about 25 nt.
  • the transcription termination sequence is one that is functional in a eukaryotic cell. In some cases, the transcription termination sequence is one that is functional in a prokaryotic cell.
  • Non-limiting examples of nucleotide sequences that can be included in a stability control sequence include sequences set forth in SEQ ID NO:683-696 and, for example, 5′-UAAUCCCACAGCCGCCAGUUCCGCUGGCGGCAUUUU-5′ (SEQ ID NO:795) (a Rho-independent trp termination site).
  • a guide RNA comprises an additional segment or segments (in some cases at the 5′ end, in some cases the 3′ end, in some cases at either the 5′ or 3′ end, in some cases embedded within the sequence (i.e., not at the 5′ and/or 3′ end), in some cases at both the 5′ end and the 3′ end, in some cases embedded and at the 5′ end and/or the 3′ end, etc).
  • a suitable additional segment can comprise a 5′ cap (e.g., a 7-methylguanylate cap (m 7 G)); a 3′ polyadenylated tail (i.e., a 3′ poly(A) tail); a ribozyme sequence (e.g.
  • a riboswitch sequence e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes
  • a sequence that forms a dsRNA duplex i.e., a hairpin
  • a sequence that targets an RNA to a subcellular location e.g., nucleus, mitochondria, chloroplasts, and the like
  • a modification or sequence that provides for tracking e.g., a direct lable (e.g., direct conjugation to a fluorescent molecule (i.e., fluorescent dye)), conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection; a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activ
  • the subject methods comprise contacting the target DNA with a donor polynucleotide, wherein the donor polynucleotide, a portion of the donor polynucleotide, a copy of the donor polynucleotide, or a portion of a copy of the donor polynucleotide integrates into the target DNA (e.g., via homology-directed repair).
  • the method does not comprise contacting the cell with a donor polynucleotide (e.g., resulting in non-homologous end-joining).
  • a donor poly nucleotide can be introduced into a target cell using any convenient technique for introducing nucleic acids into cells.
  • a polynucleotide comprising a donor sequence to be inserted is provided to the cell (e.g., the target DNA is contacted with a donor polynucleotide in addition to a Cas9 targeting complex).
  • a donor sequence or “donor polynucleotide” it is meant a nucleic acid sequence to be inserted at the cleavage site induced by a Cas9 protein.
  • a suitable donor polynucleotide can be single stranded or double stranded.
  • a donor polynucleotide is single stranded (e.g., in some cases can be referred to as an oligonucleotide), and in some cases a donor polynucleotide is double stranded (e.g., in some cases can be include two separate oligonucleotides that are hybridized).
  • the donor polynucleotide will contain sufficient homology to a genomic sequence at the cleavage site, e.g. 70%, 80%, 85%, 90%, 95%, or 100% homology with the nucleotide sequences flanking the cleavage site, e.g.
  • cleavage site within 100 bases or less (e.g., 50 bases or less of the cleavage site, e.g. within 30 bases, within 15 bases, within 10 bases, within 5 bases, or immediately flanking the cleavage site), to support homology-directed repair between it and the genomic sequence to which it bears homology.
  • nt nucleotides (e.g., 30 nt or more, 40 nt or more, 50 nt or more, 60 nt or more, 70 nt or more, 80 nt or more, 90 nt or more, 100 nt or more, 150 nt or more, 200 nt or more, etc.) of sequence homology between a donor and a genomic sequence (or any integral value between 10 and 200 nucleotides, or more) can support homology-directed repair.
  • the 5′ and/or the 3′ flanking homology arm (e.g., in some cases both of the flanking homology arms) of a donor polynucleotide can be 30 nucleotides (nt) or more in length (e.g., 40 nt or more, 50 nt or more, 60 nt or more, 70 nt or more, 80 nt or more, 90 nt or more, 100 nt or more, etc.).
  • the 5′ and/or the 3′ flanking homology arm (e.g., in some cases both of the flanking homology arms) of a donor polynucleotide can have a length in a range of from 30 nt to 500 nt (e.g., 30 nt to 400 nt, 30 nt to 350 nt, 30 nt to 300 nt, 30 nt to 250 nt, 30 nt to 200 nt, 30 nt to 150 nt, 30 nt to 100 nt, 30 nt to 90 nt, 30 nt to 80 nt, 50 nt to 400 nt, 50 nt to 350 nt, 50 nt to 300 nt, 50 nt to 250 nt, 50 nt to 200 nt, 50 nt to 150 nt, 50 nt to 100 nt, 50 nt to 90 nt, 50 nt to 80 nt
  • Donor sequences can be of any length, e.g. 10 nucleotides or more, 50 nucleotides or more, 100 nucleotides or more, 250 nucleotides or more, 500 nucleotides or more, 1000 nucleotides or more, 5000 nucleotides or more, etc.
  • the donor sequence is typically not identical to the genomic sequence that it replaces. Rather, the donor sequence may contain at least one or more single base changes, insertions, deletions, inversions or rearrangements with respect to the genomic sequence, so long as sufficient homology is present to support homology-directed repair.
  • the donor sequence comprises a non-homologous sequence flanked by two regions of homology, such that homology-directed repair between the target DNA region and the two flanking sequences results in insertion of the non-homologous sequence at the target region.
  • Donor sequences may also comprise a vector backbone containing sequences that are not homologous to the DNA region of interest and that are not intended for insertion into the DNA region of interest.
  • the homologous region(s) of a donor sequence will have at least 50% sequence identity to a genomic sequence with which recombination is desired. In certain embodiments, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or 99.9% sequence identity is present. Any value between 1% and 100% sequence identity can be present, depending upon the length of the donor polynucleotide.”
  • a donor polynucleotide is delivered to the cell (introduced into a cell) as part of viral vector (e.g., an adeno-associated virus (AAV) vector) (e.g., in combination with cell synchronization, in combination with blocking a cell at a desired phase in the cell cycle, etc., as described elsewhere in this disclosure).
  • viral vector e.g., an adeno-associated virus (AAV) vector
  • AAV DNA e.g., AAV DNA
  • donor sequence e.g., a virus, e.g., AAV, can include a DNA molecule that includes a donor polynucleotide sequence.
  • a donor polynucleotide is introduced into a cell as a virus (e.g., an AAV, e.g., the donor polynucleotide sequence is present as part of the viral DNA, e.g., AAV DNA) and the Cas9 protein and Cas9 guide RNA are delivered by a different route.
  • a donor polynucleotide is introduced into a cell as a virus (e.g., an AAV, e.g., the donor polynucleotide sequence is present as part of the viral DNA, e.g., AAV DNA) and the Cas9 protein and Cas9 guide RNA are delivered as part of a separate expression vector.
  • a donor polynucleotide is introduced into a cell as a virus (e.g., an AAV, e.g., the donor polynucleotide sequence is present as part of the viral DNA, e.g., AAV DNA) and the Cas9 protein and Cas9 guide RNA are delivered as part of a ribonucleoprotein complex (RNP) (e.g., described in more detail below).
  • a virus e.g., an AAV
  • the donor polynucleotide sequence is present as part of the viral DNA, e.g., AAV DNA
  • Cas9 protein and Cas9 guide RNA are delivered as part of a ribonucleoprotein complex (RNP) (e.g., described in more detail below).
  • RNP ribonucleoprotein complex
  • a donor polynucleotide is introduced into a cell as a virus (e.g., an AAV, e.g., the donor polynucleotide sequence is present as part of the viral DNA, e.g., AAV DNA),
  • the Cas9 guide RNA is delivered as either an RNA or DNA encoding the RNA, and
  • a Cas9 protein is delivered as a protein or as a nucleic acid encoding the protein (e.g., RNA or DNA).
  • a recombinant viral vector comprising a donor polynucleotide is introduced into a cell before the Cas9-guide RNA RNP is introduced into the cell.
  • a recombinant viral vector e.g., a recombinant AAV vector
  • a recombinant viral vector comprising a donor polynucleotide is introduced into a cell from 2 hours to 72 hours (e.g., from 2 hours to 4 hours, from 4 hours to 8 hours, from 8 hours to 12 hours, from 12 hours to 24 hours, from 24 hours to 48 hours, or from 48 hours to 72 hours) before the Cas9-guide RNA RNP is introduced into the cell.
  • a subject method comprises: a) blocking a cell at a desired phase in the cell cycle; b) contacting the cell with a recombinant viral vector (e.g., a recombinant AAV vector) comprising a donor polynucleotide; and c) contacting the cell with a Cas9-guide RNA RNP.
  • a recombinant viral vector e.g., a recombinant AAV vector
  • kits and compositions that include (i) a viral vector (e.g., a viral DNA, a virion, etc.)(e.g., an AAV vector) that includes the donor polynucleotide (i.e., the viral DNA includes the donor sequence); (ii) a Cas9 protein or nucleic acid encoding the protein; (iii) a Cas9 guide RNA or a DNA encoding the Cas9 guide RNA; and/or (iv) a cell cycle blocking composition (e.g., a composition that includes at least one of nocodazole, hydroxyurea; colchicine; demecolcine (colcemid); lovastatin; mimosine; thymidine; aphidicolin; latrunculin A; and latrunculin B).
  • a viral vector e.g., a viral DNA, a virion, etc.
  • an AAV vector that includes the donor polynucleotide (i.e., the
  • kits and compositions that include (i) a viral vector (e.g., a viral DNA, a virion, etc.)(e.g., an AAV vector) that includes the donor polynucleotide (i.e., the viral DNA includes the donor sequence); (ii) an RNP that includes a Cas9 protein and a Cas9 guide RNA; and/or (iii) a cell cycle blocking composition (e.g., a composition that includes at least one of nocodazole, hydroxyurea; colchicine; demecolcine (colcemid); lovastatin; mimosine; thymidine; aphidicolin; latrunculin A; and latrunculin B).
  • a viral vector e.g., a viral DNA, a virion, etc.
  • an AAV vector that includes the donor polynucleotide (i.e., the viral DNA includes the donor sequence)
  • an RNP that includes a Cas9 protein and a
  • a suitable guide RNA and a suitable Cas9 protein form a Cas9 targeting complex.
  • the guide RNA provides target specificity to the complex by comprising a nucleotide sequence that is complementary to a sequence (the target site) of a target nucleic acid (as noted above).
  • the Cas9 protein of the complex provides the site-specific activity.
  • the Cas9 protein is guided to a target site within a target nucleic acid sequence (e.g. a chromosomal sequence or an extrachromosomal sequence, e.g. an episomal sequence, a minicircle sequence, a mitochondrial sequence, a chloroplast sequence, etc.) by virtue of its association with the protein-binding segment of the guide RNA (described above).
  • a suitable Cas9 protein can bind and/or modify (e.g., cleave, methylate, demethylate, etc.) a target nucleic acid and/or a polypeptide associated with target nucleic acid (e.g., methylation or acetylation of a histone tail).
  • a Cas9 protein is also referred to herein as a “site-directed polypeptide.”
  • the Cas9 protein is a naturally-occurring polypeptide (e.g, naturally occurs in bacterial and/or archaeal cells). In other cases, the Cas9 protein is not a naturally-occurring polypeptide (e.g., the Cas9 protein is a variant Cas9 protein, a chimeric polypeptide as discussed below, and the like).
  • Exemplary Cas9 proteins are set forth in SEQ ID NOs: 1-259, and 795-1346 as a non-limiting and non-exhaustive list of Cas9 endonucleases.
  • Naturally occurring Cas9 proteins bind a guide RNA, are thereby directed to a specific sequence within a target nucleic acid (a target site), and cleave the target nucleic acid (e.g., cleave dsDNA to generate a double strand break, cleave ssDNA, cleave ssRNA, etc.).
  • a suitable Cas9 protein comprises two portions, an RNA-binding portion and an activity portion. An RNA-binding portion interacts with a guide RNA.
  • An activity portion exhibits site-directed enzymatic activity (e.g., nuclease activity, activity for DNA and/or RNA methylation, activity for DNA and/or RNA cleavage, activity for histone acetylation, activity for histone methylation, activity for RNA modification, activity for RNA-binding, activity for RNA splicing etc.).
  • site-directed enzymatic activity e.g., nuclease activity, activity for DNA and/or RNA methylation, activity for DNA and/or RNA cleavage, activity for histone acetylation, activity for histone methylation, activity for RNA modification, activity for RNA-binding, activity for RNA splicing etc.
  • the activity portion exhibits reduced nuclease activity relative to the corresponding portion of a wild type Cas9 protein.
  • the activity portion is enzymatically inactive.
  • the activity portion is a general term meant to encompass all parts of the Cas9 protein that exhibits an activity
  • Cas9 is known to have two domains that function as nuclease domains, and the domains are separated from each other in the primary amino acid sequence, the domains can be collectively referred to as an activity domain (activity portion).
  • the Cas9 polypeptide has an activity portion that is provided by a heterologous sequence (e.g., a methylation activity).
  • Assays to determine whether a protein has an RNA-binding portion interacts with a subject guide RNA can be any convenient binding assay that tests for binding between a protein and a nucleic acid. Exemplary binding assays will be known to one of ordinary skill in the art and can be found for example in U.S.
  • patent applications 20140068797, 20140189896, 20140179006, 20140170753, 20140179770, 20140186958, 20140186919, 20140186843; international applications: WO2013176772, WO2013141680, WO2013142578, WO2014065596, WO2014089290, WO2014099744, WO2014099750, WO2014104878, WO2014093718, WO2014093622, WO2014093655, WO2014093701, WO2014093712, WO2014093635, WO2014093595, WO2014093694, and WO2014093661; and U.S. Pat. Nos.
  • Suitable assays include, without limitation, binding assays (e.g., gel shift assays) that include adding a guide RNA and a Cas9 protein to a target nucleic acid.
  • binding assays e.g., gel shift assays
  • Assays to determine whether a protein has an activity portion can be any convenient nucleic acid cleavage assay that tests for nucleic acid cleavage. Exemplary cleavage assays can be found in U.S.
  • patent applications 20140068797, 20140189896, 20140179006, 20140170753, 20140179770, 20140186958, 20140186919, 20140186843; international applications: WO2013176772, WO2013141680, WO2013142578, WO2014065596, WO2014089290, WO2014099744, WO2014099750, WO2014104878, WO2014093718, WO2014093622, WO2014093655, WO2014093701, WO2014093712, WO2014093635, WO2014093595, WO2014093694, and WO2014093661; and U.S. Pat. Nos. 8,697,359, 8,771,945.
  • Suitable assays can include cleavage assays that include adding a guide RNA and a Cas9 protein to a target nucleic acid.
  • a PAM-mer is also added (e.g., in some cases when the target nucleic acid is a single stranded nucleic acid).
  • a subject Cas9 protein (e.g., a chimeric Cas9 protein) has enzymatic activity that modifies target nucleic acid (e.g., nuclease activity, methyltransferase activity, demethylase activity, DNA repair activity, DNA damage activity, deamination activity, dismutase activity, alkylation activity, depurination activity, oxidation activity, pyrimidine dimer forming activity, integrase activity, transposase activity, recombinase activity, polymerase activity, ligase activity, helicase activity, photolyase activity or glycosylase activity).
  • target nucleic acid e.g., nuclease activity, methyltransferase activity, demethylase activity, DNA repair activity, DNA damage activity, deamination activity, dismutase activity, alkylation activity, depurination activity, oxidation activity, pyrimidine dimer forming activity, integrase activity, trans
  • a suitable Cas9 protein (a chimeric Cas9 protein) has enzymatic activity that modifies a polypeptide (e.g., a histone) associated with target nucleic acid (e.g., methyltransferase activity, demethylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase activity, ubiquitin ligase activity, deubiquitinating activity, adenylation activity, deadenylation activity, SUMOylating activity, deSUMOylating activity, ribosylation activity, deribosylation activity, myristoylation activity or demyristoylation activity).
  • a polypeptide e.g., a histone
  • target nucleic acid e.g., methyltransferase activity, demethylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphata
  • Cas9 orthologs from a wide variety of species have been identified and the proteins share only a few identical amino acids.
  • the identified Cas9 orthologs have the same domain architecture with a central HNH endonuclease domain and a split RuvC/RNaseH domain.
  • Cas9 proteins share 4 key motifs with a conserved architecture. Motifs 1, 2, and 4 are RuvC like motifs while motif 3 is an HNH-motif.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 60% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 99% or more or 100% amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence (e.g., SEQ ID NOs:260-263, respectively, as depicted in Table 1), or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346. Additional Cas9 protein sequences can be found in U.S.
  • patent applications 20140068797, 20140189896, 20140179006, 20140170753, 20140179770, 20140186958, 20140186919, 20140186843; international applications: WO2013176772, WO2013141680, WO2013142578, WO2014065596, WO2014089290, WO2014099744, WO2014099750, WO2014104878, WO2014093718, WO2014093622, WO2014093655, WO2014093701, WO2014093712, WO2014093635, WO2014093595, WO2014093694, and WO2014093661; and U.S. Pat. Nos. 8,697,359, 8,771,945.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 60% or more amino acid sequence identity to motifs 1-4 of the SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 70% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 75% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 80% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 85% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 90% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 95% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 99% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 100% amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequences set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable Cas9 protein comprises an amino acid sequence having 60% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 99% or more or 100% amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 60% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 70% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 75% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 80% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 85% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 90% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 95% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 99% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a suitable Cas9 protein comprises an amino acid sequence having 100% amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a Cas9 protein or as part of a chimeric Cas9 protein of the subject methods.
  • a Cas9 protein comprises 4 motifs (as listed in Table 1), at least one with (or each with) amino acid sequences having 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 99% or more or 100% amino acid sequence identity to each of the 4 motifs listed in Table 1(SEQ ID NOs:260-263), or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Cas9 protein encompasses the term “variant Cas9 protein”; and the term “variant Cas9 protein” encompasses the term “chimeric Cas9 protein.”
  • a method of the present disclosure involves use of a variant Cas9 protein.
  • a variant Cas9 polypeptide has an amino acid sequence that is different by one amino acid (e.g., has a deletion, insertion, substitution, fusion) when compared to the amino acid sequence of a wild type Cas9 protein.
  • the variant Cas9 polypeptide has an amino acid change (e.g., deletion, insertion, or substitution) that reduces the nuclease activity of the Cas9 polypeptide.
  • the variant Cas9 polypeptide has less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, or less than 1% of the nuclease activity of the corresponding wild-type Cas9 protein. In some cases, the variant Cas9 protein has no substantial nuclease activity.
  • a subject Cas9 protein is a variant Cas9 protein that has no substantial nuclease activity, it can be referred to as “dCas9.”
  • a variant Cas9 protein has reduced nuclease activity.
  • a variant Cas9 protein suitable for use in a binding method of the present disclosure exhibits less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 1%, or less than about 0.1%, of the endonuclease activity of a wild-type Cas9 protein, e.g., a wild-type Cas9 protein comprising an amino acid sequence as set forth in SEQ ID NO:8.
  • a variant Cas9 protein can cleave the complementary strand of a target DNA but has reduced ability to cleave the non-complementary strand of a double stranded target DNA.
  • the variant Cas9 protein can have a mutation (amino acid substitution) that reduces the function of the RuvC domain.
  • a variant Cas9 protein has a D10A (aspartate to alanine at amino acid position 10 of SEQ ID NO:8 mutation (or the corresponding mutation of any of the proteins presented in SEQ ID NOs:1-256 and 795-1346) and can therefore cleave the complementary strand of a double stranded target DNA but has reduced ability to cleave the non-complementary strand of a double stranded target DNA (thus resulting in a single strand break (SSB) instead of a double strand break (DSB) when the variant Cas9 protein cleaves a double stranded target nucleic acid) (see, for example, Jinek et al., Science. 2012 Aug. 17; 337(6096):816-21).
  • D10A aspartate to alanine at amino acid position 10 of SEQ ID NO:8 mutation (or the corresponding mutation of any of the proteins presented in SEQ ID NOs:1-256 and 795-1346) and can therefore cleave
  • a variant Cas9 protein can cleave the non-complementary strand of a double stranded target DNA but has reduced ability to cleave the complementary strand of the target DNA.
  • the variant Cas9 protein can have a mutation (amino acid substitution) that reduces the function of the HNH domain (RuvC/HNH/RuvC domain motifs).
  • the variant Cas9 protein has an H840A (histidine to alanine at amino acid position 840) mutation (or the corresponding mutation of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) and can therefore cleave the non-complementary strand of the target DNA but has reduced ability to cleave the complementary strand of the target DNA (thus resulting in a SSB instead of a DSB when the variant Cas9 protein cleaves a double stranded target DNA).
  • H840A histidine to alanine at amino acid position 840
  • Such a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • a variant Cas9 protein has a reduced ability to cleave both the complementary and the non-complementary strands of a double stranded target DNA.
  • the variant Cas9 protein harbors both the D10A and the H840A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave both the complementary and the non-complementary strands of a double stranded target DNA.
  • Such a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors W476A and W1126A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • W476A and W1126A mutations or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346
  • Such a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors P475A, W476A, N477A, D1125A, W1126A, and D1127A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors H840A, W476A, and W1126A, mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors H840A, D10A, W476A, and W1126A, mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors, H840A, P475A, W476A, N477A, D1125A, W1126A, and D1127A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein harbors D10A, H840A, P475A, W476A, N477A, D1125A, W1126A, and D1127A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346) such that the polypeptide has a reduced ability to cleave a target DNA.
  • a Cas9 protein has a reduced ability to cleave a target DNA (e.g., a single stranded target DNA) but retains the ability to bind a target DNA (e.g., a single stranded target DNA).
  • the variant Cas9 protein when a variant Cas9 protein harbors W476A and W1126A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346); or when the variant Cas9 protein harbors P475A, W476A, N477A, D1125A, W1126A, and D1127A mutations (or the corresponding mutations of any of the proteins set forth as SEQ ID NOs:1-256 and 795-1346), the variant Cas9 protein does not bind efficiently to a PAM sequence. Thus, in some such cases, when such a variant Cas9 protein is used in a method of binding, the method need not include a PAM-mer.
  • the method can include a guide RNA, but the method can be performed in the absence of a PAM-mer (and the specificity of binding is therefore provided by the targeting segment of the guide RNA).
  • residues D10, G12, G17, E762, H840, N854, N863, H982, H983, A984, D986, and/or A987 can be altered (i.e., substituted). Also, mutations other than alanine substitutions are suitable.
  • a variant Cas9 protein that has reduced catalytic activity e.g., when a Cas9 protein has a D10, G12, G17, E762, H840, N854, N863, H982, H983, A984, D986, and/or a A987 mutation, e.g., D10A, G12A, G17A, E762A, H840A, N854A, N863A, H982A, H983A, A984A, and/or D986A), the variant Cas9 protein can still bind to target DNA in a site-specific manner (because it is still guided to a target DNA sequence by a guide RNA) as long as it retains the ability to interact with the guide RNA.
  • the variant Cas9 protein can still bind to target DNA in a site-specific manner (because it is still guided to a target DNA sequence by a guide RNA) as long as it retains the ability to interact with the guide RNA.
  • Table 1 lists 4 motifs that are present in Cas9 sequences from various species. The amino acids listed here are from the Cas9 from S . pyogenes (SEQ ID NO: 8. Motif # Motif Amino acids (residue #s) Highly conserved 1 RuvC-like I IGLDIGTNSVGWAVI (7-21) D10, G12, G17 (SEQ ID NO: 260) 2 RuvC-like II IVIEMARE (759-766) E762 (SEQ ID NO: 261) 3 HNH-motif DVDHIVPQSFLKDDSIDNKVLTRSDKN H840, N854, N863 (837-863) (SEQ ID NO: 262) 4 RuvC-like II HHAHDAYL (982-989) H982, H983, A984, (SEQ ID NO: 263) D986, A987
  • a variant Cas9 protein can have the same parameters for sequence identity as described above for Cas9 proteins.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 60% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 99% or more or 100% amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 60% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 70% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 75% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 80% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 85% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 90% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 95% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 99% or more amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 4 motifs, each of motifs 1-4 having 100% amino acid sequence identity to motifs 1-4 of the Cas9 amino acid sequence set forth in SEQ ID NOs:260-263, respectively, as depicted in Table 1, or to the corresponding portions in any of the amino acid sequences set forth in SEQ ID NOs:1-256 and 795-1346.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 60% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 99% or more, or 100% amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 60% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 70% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 75% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 80% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 85% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 90% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 95% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 99% or more amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a suitable variant Cas9 protein comprises an amino acid sequence having 100% amino acid sequence identity to amino acids 7-166 or 731-1003 of the Cas9 amino acid sequence set forth in SEQ ID NO:8, or to the corresponding portions in any of the amino acid sequences set forth as SEQ ID NOs:1-256 and 795-1346.
  • Any Cas9 protein as defined above can be used as a variant Cas9 protein or as part of a chimeric variant Cas9 protein of the subject methods.
  • a variant Cas9 protein is a chimeric Cas9 protein (also referred to herein as a fusion polypeptide, e.g., a “Cas9 fusion polypeptide”).
  • a Cas9 fusion polypeptide can bind and/or modify a target DNA (e.g., cleave, methylate, demethylate, etc.) and/or a polypeptide associated with target DNA (e.g., methylation, acetylation, etc., of, for example, a histone tail).
  • a Cas9 fusion polypeptide is a variant Cas9 protein by virtue of differing in sequence from a wild type Cas9 protein.
  • a Cas9 fusion polypeptide is a Cas9 protein (e.g., a wild type Cas9 protein, a variant Cas9 protein, a variant Cas9 protein with reduced nuclease activity (as described above), and the like) fused to a covalently linked heterologous polypeptide (also referred to as a “fusion partner”).
  • a Cas9 fusion polypeptide is a variant Cas9 protein with reduced nuclease activity (e.g., cleaves the complementary strand of the target DNA but does not cleave the non-complementary strand; cleaves the non-complementary strand of the target DNA but does not cleave the complementary strand; does not cleave either the complementary strand for the non-complementary strand if target DNA (dCas9); as described in more detail in the section related to variant Cas9 polypeptides) fused to a covalently linked heterologous polypeptide.
  • dCas9 target DNA
  • the heterologous polypeptide exhibits (and therefore provides for) an activity (e.g., an enzymatic activity) that will also be exhibited by the Cas9 fusion polypeptide (e.g., methyltransferase activity, acetyltransferase activity, kinase activity, ubiquitinating activity, etc.).
  • an activity e.g., an enzymatic activity
  • a method of binding e.g., where the Cas9 protein is a variant Cas9 protein having a fusion partner (i.e., having a heterologous polypeptide) with an activity (e.g., an enzymatic activity) that modifies the target DNA
  • the method can also be considered to be a method of modifying the target DNA.
  • a method of binding a target DNA can result in modification of the target DNA.
  • a method of binding a target DNA can be a method of modifying the target DNA.
  • a Cas9 protein has a heterologous sequence that provides for subcellular localization (i.e., the heterologous sequence is a subcellular localization sequence (e.g., a nuclear localization signal (NLS) for targeting to the nucleus, a sequence to keep the fusion protein out of the nucleus (e.g., a nuclear export sequence (NES), a sequence to keep the fusion protein retained in the cytoplasm, a mitochondrial localization signal for targeting to the mitochondria, a chloroplast localization signal for targeting to a chloroplast, an ER retention signal, and the like).
  • a subcellular localization sequence e.g., a nuclear localization signal (NLS) for targeting to the nucleus
  • a sequence to keep the fusion protein out of the nucleus e.g., a nuclear export sequence (NES)
  • NES nuclear export sequence
  • a sequence to keep the fusion protein retained in the cytoplasm e.g., a mitochondrial localization signal for targeting to the mitochondria,
  • a variant Cas9 does not include a NLS so that the protein is not targeted to the nucleus (which can be advantageous, e.g., when the target DNA is an RNA that is present in the cyosol).
  • the heterologous sequence can provide a tag (i.e., the heterologous sequence is a detectable label) for ease of tracking and/or purification (e.g., a fluorescent protein, e.g., green fluorescent protein (GFP), YFP, RFP, CFP, mCherry, tdTomato, and the like; a histidine tag, e.g., a 6 ⁇ His tag; a hemagglutinin (HA) tag; a FLAG tag; a Myc tag; and the like).
  • GFP green fluorescent protein
  • YFP green fluorescent protein
  • RFP RFP
  • CFP CFP
  • mCherry mCherry
  • tdTomato e.g., a histidine tag
  • the heterologous sequence can provide for increased or decreased stability (i.e., the heterologous sequence is a stability control peptide, e.g., a degron, which in some cases is controllable (e.g., a temperature sensitive or drug controllable degron sequence, see below).
  • a stability control peptide e.g., a degron
  • controllable e.g., a temperature sensitive or drug controllable degron sequence, see below.
  • the heterologous sequence can provide for increased or decreased transcription from the target DNA (i.e., the heterologous sequence is a transcription modulation sequence, e.g., a transcription factor/activator or a fragment thereof, a protein or fragment thereof that recruits a transcription factor/activator, a transcription repressor or a fragment thereof, a protein or fragment thereof that recruits a transcription repressor, a small molecule/drug-responsive transcription regulator, etc.).
  • a transcription modulation sequence e.g., a transcription factor/activator or a fragment thereof, a protein or fragment thereof that recruits a transcription factor/activator, a transcription repressor or a fragment thereof, a protein or fragment thereof that recruits a transcription repressor, a small molecule/drug-responsive transcription regulator, etc.
  • the heterologous sequence can provide a binding domain (i.e., the heterologous sequence is a protein binding sequence, e.g., to provide the ability of a Cas9 fusion polypeptide to bind to another protein of interest, e.g., a DNA or histone modifying protein, a transcription factor or transcription repressor, a recruiting protein, an RNA modification enzyme, an RNA-binding protein, a translation initiation factor, an RNA splicing factor, etc.).
  • a heterologous nucleic acid sequence may be linked to another nucleic acid sequence (e.g., by genetic engineering) to generate a chimeric nucleotide sequence encoding a chimeric polypeptide.
  • Suitable fusion partners that provide for increased or decreased stability include, but are not limited to degron sequences.
  • Degrons are readily understood by one of ordinary skill in the art to be amino acid sequences that control the stability of the protein of which they are part. For example, the stability of a protein comprising a degron sequence is controlled in part by the degron sequence.
  • a suitable degron is constitutive such that the degron exerts its influence on protein stability independent of experimental control (i.e., the degron is not drug inducible, temperature inducible, etc.)
  • the degron provides the variant Cas9 protein with controllable stability such that the variant Cas9 protein can be turned “on” (i.e., stable) or “off” (i.e., unstable, degraded) depending on the desired conditions.
  • the variant Cas9 protein may be functional (i.e., “on”, stable) below a threshold temperature (e.g., 42° C., 41° C., 40° C., 39° C., 38° C., 37° C., 36° C., 35° C., 34° C., 33° C., 32° C., 31° C., 30° C., etc.) but non-functional (i.e., “off”, degraded) above the threshold temperature.
  • a threshold temperature e.g., 42° C., 41° C., 40° C., 39° C., 38° C., 37° C., 36° C., 35° C., 34° C., 33° C., 32° C., 31° C., 30° C., etc.
  • non-functional i.e., “off”, degraded
  • the degron is a drug inducible degron
  • the presence or absence of drug can switch the protein from an “off” (i.e., unstable) state to an “on” (i.e., stable) state or vice versa.
  • An exemplary drug inducible degron is derived from the FKBP12 protein. The stability of the degron is controlled by the presence or absence of a small molecule that binds to the degron.
  • suitable degrons include, but are not limited to those degrons controlled by Shield-1, DHFR, auxins, and/or temperature.
  • suitable degrons are known in the art (e.g., Dohmen et al., Science, 1994. 263(5151): p. 1273-1276: Heat-inducible degron: a method for constructing temperature-sensitive mutants; Schoeber et al., Am J Physiol Renal Physiol. 2009 January; 296(1):F204-11: Conditional fast expression and function of multimeric TRPVS channels using Shield-1; Chu et al., Bioorg Med Chem Lett. 2008 Nov.
  • Exemplary degron sequences have been well-characterized and tested in both cells and animals.
  • Cas9 e.g., wild type Cas9; variant Cas9; variant Cas9 with reduced nuclease activity, e.g., dCas9; and the like
  • Any of the fusion partners described herein can be used in any desirable combination.
  • a Cas9 fusion protein i.e., a chimeric Cas9 protein
  • a suitable reporter protein for use as a fusion partner for a Cas9 protein includes, but is not limited to, the following exemplary proteins (or functional fragment thereof): his3, ⁇ -galactosidase, a fluorescent protein (e.g., GFP, RFP, YFP, cherry, tomato, etc., and various derivatives thereof), luciferase, ⁇ -glucuronidase, and alkaline phosphatase.
  • a Cas9 fusion protein comprises one or more (e.g. two or more, three or more, four or more, or five or more) heterologous sequences.
  • Suitable fusion partners include, but are not limited to, a polypeptide that provides for methyltransferase activity, demethylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase activity, ubiquitin ligase activity, deubiquitinating activity, adenylation activity, deadenylation activity, SUMOylating activity, deSUMOylating activity, ribosylation activity, deribosylation activity, myristoylation activity, or demyristoylation activity, any of which can be directed at modifying nucleic acid directly (e.g., methylation of DNA or RNA) or at modifying a nucleic acid-associated polypeptide (e.g., a histone, a DNA binding protein, and RNA binding protein, and the like).
  • a nucleic acid-associated polypeptide e.g., a histone, a DNA binding protein, and RNA binding protein, and
  • fusion partners include, but are not limited to boundary elements (e.g., CTCF), proteins and fragments thereof that provide periphery recruitment (e.g., Lamin A, Lamin B, etc.), and protein docking elements (e.g., FKBP/FRB, Pill/Aby1, etc.).
  • boundary elements e.g., CTCF
  • proteins and fragments thereof that provide periphery recruitment e.g., Lamin A, Lamin B, etc.
  • protein docking elements e.g., FKBP/FRB, Pill/Aby1, etc.
  • Examples of various additional suitable fusion partners (or fragments thereof) for a subject variant Cas9 protein include, but are not limited to those described in the PCT patent applications: WO2010075303, WO2012068627, and WO2013155555 which are hereby incorporated by reference in their entirety.
  • Suitable fusion partners include, but are not limited to, a polypeptide that provides an activity that indirectly increases transcription by acting directly on the target DNA or on a polypeptide (e.g., a histone, a DNA-binding protein, an RNA-binding protein, an RNA editing protein, etc.) associated with the target DNA.
  • a polypeptide e.g., a histone, a DNA-binding protein, an RNA-binding protein, an RNA editing protein, etc.
  • Suitable fusion partners include, but are not limited to, a polypeptide that provides for methyltransferase activity, demethylase activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase activity, ubiquitin ligase activity, deubiquitinating activity, adenylation activity, deadenylation activity, SUMOylating activity, deSUMOylating activity, ribosylation activity, deribosylation activity, myristoylation activity, or demyristoylation activity.
  • Additional suitable fusion partners include, but are not limited to, a polypeptide that directly provides for increased transcription and/or translation of a target DNA (e.g., a transcription activator or a fragment thereof, a protein or fragment thereof that recruits a transcription activator, a small molecule/drug-responsive transcription and/or translation regulator, a translation-regulating protein, etc.).
  • a target DNA e.g., a transcription activator or a fragment thereof, a protein or fragment thereof that recruits a transcription activator, a small molecule/drug-responsive transcription and/or translation regulator, a translation-regulating protein, etc.
  • Non-limiting examples of fusion partners to accomplish increased or decreased transcription include, e.g., transcription activator and transcription repressor domains (e.g., the Krüppel associated box (KRAB or SKD); the Mad mSIN3 interaction domain (SID); the ERF repressor domain (ERD), etc).
  • transcription activator and transcription repressor domains e.g., the Krüppel associated box (KRAB or SKD); the Mad mSIN3 interaction domain (SID); the ERF repressor domain (ERD), etc.
  • a Cas9 fusion protein is targeted by the guide RNA to a specific location (i.e., sequence) in the target DNA and exerts locus-specific regulation such as blocking RNA polymerase binding to a promoter (which selectively inhibits transcription activator function), and/or modifying the local chromatin status (e.g., when a fusion sequence is used that modifies the target DNA or modifies a polypeptide associated with the target DNA).
  • locus-specific regulation such as blocking RNA polymerase binding to a promoter (which selectively inhibits transcription activator function), and/or modifying the local chromatin status (e.g., when a fusion sequence is used that modifies the target DNA or modifies a polypeptide associated with the target DNA).
  • the changes are transient (e.g., transcription repression or activation).
  • the changes are inheritable (e.g., when epigenetic modifications are made to the target DNA or to proteins associated with the target DNA, e.g., nucleosomal histone
  • the heterologous sequence can be fused to the C-terminus of the Cas9 protein. In some embodiments, the heterologous sequence can be fused to the N-terminus of the Cas9 protein. In some embodiments, the heterologous sequence can be fused to an internal portion (i.e., a portion other than the N- or C-terminus) of the Cas9 protein.
  • a Cas9 protein e.g., a wild type Cas9, a variant Cas9, a variant Cas9 with reduced nuclease activity, etc.
  • a Cas9 protein can be linked to a fusion partner via a peptide spacer.
  • Methods of the present disclosure include contacting a target cell (or contacting enriched cells of a cell population) with a Cas9 targeting complex.
  • Contacting can include introducing into a cell at least one of: (a) a single guide RNA, (b) a DNA polynucleotide encoding a single guide RNA, (c) a targeter-RNA, (d) a DNA polynucleotide encoding a targeter-RNA, (e) an activator-RNA, (f) a DNA polynucleotide encoding an activator-RNA, (g) a Cas9 protein, and (h) a nucleic acid encoding a Cas9 protein.
  • the target cell already has a nucleic acid encoding a Cas9 protein the one performing the method may only need to introduce the guide RNA component(s) of the Cas9 targeting complex.
  • a component of a Cas9 targeting complex (a targeter-RNA, an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein) is provided as a nucleic acid encoding the component.
  • a subject nucleic acid is an expression vector, e.g., a recombinant expression vector.
  • a subject method involves contacting a target DNA (e.g., via introducing into a target cell or a population of cells) with a nucleic acid encoding a targeter-RNA, an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein.
  • a target DNA e.g., via introducing into a target cell or a population of cells
  • a targeter-RNA e.g., an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein.
  • a cell comprising a target DNA is in vitro and/or ex vivo. In some embodiments a cell comprising a target DNA is in vivo.
  • Suitable nucleic acids comprising nucleotide sequences encoding a targeter-RNA, an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein include expression vectors (e.g., recombinant expression vectors).
  • the recombinant expression vector is a viral construct, e.g., a recombinant adeno-associated virus construct (see, e.g., U.S. Pat. No. 7,078,387), a recombinant adenoviral construct, a recombinant lentiviral construct, a recombinant retroviral construct, etc.
  • a viral construct e.g., a recombinant adeno-associated virus construct (see, e.g., U.S. Pat. No. 7,078,387), a recombinant adenoviral construct, a recombinant lentiviral construct, a recombinant retroviral construct, etc.
  • Suitable expression vectors include, but are not limited to, viral vectors (e.g. viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., Invest Opthalmol Vis Sci 35:2543 2549, 1994; Borras et al., Gene Ther 6:515 524, 1999; Li and Davidson, PNAS 92:7700 7704, 1995; Sakamoto et al., H Gene Ther 5:1088 1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655); adeno-associated virus (see, e.g., Ali et al., Hum Gene Ther 9:81 86, 1998, Flannery et al., PNAS 94:6916 6921, 1997; Bennett et al., Invest Opthalmol Vis
  • a retroviral vector e.g., Murine Leukemia Virus
  • Suitable expression vectors are known to those of skill in the art, and many are commercially available.
  • the following vectors are provided by way of example; for eukaryotic host cells: pXT1, pSG5 (Stratagene), pSVK3, pBPV, pMSG, and pSVLSV40 (Pharmacia).
  • any other vector may be used so long as it is compatible with the host cell.
  • any of a number of suitable transcription and translation control elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. may be used in the expression vector (see e.g., Bitter et al. (1987) Methods in Enzymology, 153:516-544).
  • a nucleotide sequence encoding a targeter-RNA, an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein is operably linked to a control element, e.g., a transcriptional control element, such as a promoter (e.g. a promoter functional in a eukaryotic cell).
  • a nucleotide sequence encoding a component of a Cas9 targeting complex is operably linked to multiple control elements that allow expression of the nucleotide sequence encoding a PAM-mer, and/or a guide RNA and/or a Cas9 protein in both prokaryotic and eukaryotic cells.
  • eukaryotic promoters include those from cytomegalovirus (CMV) immediate early, herpes simplex virus (HSV) thymidine kinase, early and late SV40, long terminal repeats (LTRs) from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • the expression vector may also contain a ribosome binding site for translation initiation and a transcription terminator.
  • the expression vector may also include appropriate sequences for amplifying expression.
  • the expression vector may also include nucleotide sequences encoding protein tags (e.g., 6 ⁇ His tag, hemagglutinin tag, green fluorescent protein, etc.) that are fused to the Cas9 protein, thus resulting in a chimeric polypeptide.
  • protein tags e.g., 6 ⁇ His tag, hemagglutinin tag, green fluorescent protein, etc.
  • a nucleotide sequence encoding a component of a Cas9 targeting complex is operably linked to an inducible promoter In some embodiments, a nucleotide sequence encoding a component of a Cas9 targeting complex is operably linked to a constitutive promoter.
  • a promoter can be a constitutively active promoter (i.e., a promoter that is constitutively in an active/“ON” state), it may be an inducible promoter (i.e., a promoter whose state, active/“ON” or inactive/“OFF”, is controlled by an external stimulus, e.g., the presence of a particular temperature, compound, or protein.), it may be a spatially restricted promoter (i.e., transcriptional control element, enhancer, etc.)(e.g., tissue specific promoter, cell type specific promoter, etc.), and it may be a temporally restricted promoter (i.e., the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process, e.g., hair follicle cycle in mice).
  • a constitutively active promoter i.e., a promoter that is constitutively in an active/“ON” state
  • it may be an inducible promote
  • Suitable promoters can be derived from viruses and can therefore be referred to as viral promoters, or they can be derived from any organism, including prokaryotic or eukaryotic organisms. Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • RNA polymerase e.g., pol I, pol II, pol III
  • Exemplary promoters include, but are not limited to the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6) (Miyagishi et al., Nature Biotechnology 20, 497-500 (2002)), an enhanced U6 promoter (e.g., Xia et al., Nucleic Acids Res. 2003 Sep.
  • LTR mouse mammary tumor virus long terminal repeat
  • Ad MLP adenovirus major late promoter
  • HSV herpes simplex virus
  • CMV cytomegalovirus
  • CMVIE CMV immediate early promoter region
  • RSV rous sarcoma virus
  • H1 promoter H1 promoter 1; 31(17)
  • U6 promoters are useful for expression non-coding RNAs (e.g., targeter-RNAs, activator-RNAs, single guide RNAs) in eukaryotic cells.
  • inducible promoters include, but are not limited to T7 RNA polymerase promoter, T3 RNA polymerase promoter, Isopropyl-beta-D-thiogalactopyranoside (IPTG)-regulated promoter, lactose induced promoter, heat shock promoter, Tetracycline-regulated promoter, Steroid-regulated promoter, Metal-regulated promoter, estrogen receptor-regulated promoter, etc.
  • Inducible promoters can therefore be regulated by molecules including, but not limited to, doxycycline; RNA polymerase, e.g., T7 RNA polymerase; an estrogen receptor; an estrogen receptor fusion; etc.
  • the promoter is a spatially restricted promoter (i.e., cell type specific promoter, tissue specific promoter, etc.) such that in a multi-cellular organism, the promoter is active (i.e., “ON”) in a subset of specific cells.
  • spatially restricted promoters may also be referred to as enhancers, transcriptional control elements, control sequences, etc.
  • any convenient spatially restricted promoter may be used and the choice of suitable promoter (e.g., a brain specific promoter, a promoter that drives expression in a subset of neurons, a promoter that drives expression in the germline, a promoter that drives expression in the lungs, a promoter that drives expression in muscles, a promoter that drives expression in islet cells of the pancreas, etc.) will depend on the organism.
  • various spatially restricted promoters are known for plants, flies, worms, mammals, mice, etc.
  • a spatially restricted promoter can be used to regulate the expression of a nucleic acid encoding a subject Cas9 protein in a wide variety of different tissues and cell types, depending on the organism.
  • Some spatially restricted promoters are also temporally restricted such that the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process (e.g., hair follicle cycle in mice).
  • spatially restricted promoters include, but are not limited to, neuron-specific promoters, adipocyte-specific promoters, cardiomyocyte-specific promoters, smooth muscle-specific promoters, photoreceptor-specific promoters, etc.
  • Neuron-specific spatially restricted promoters include, but are not limited to, a neuron-specific enolase (NSE) promoter (see, e.g., EMBL HSENO2, X51956); an aromatic amino acid decarboxylase (AADC) promoter; a neurofilament promoter (see, e.g., GenBank HUMNFL, L04147); a synapsin promoter (see, e.g., GenBank HUMSYNIB, M55301); a thy-1 promoter (see, e.g., Chen et al. (1987) Cell 51:7-19; and Llewellyn, et al. (2010) Nat. Med.
  • NSE neuron-specific enolase
  • AADC aromatic amino acid decarboxylase
  • Adipocyte-specific spatially restricted promoters include, but are not limited to aP2 gene promoter/enhancer, e.g., a region from ⁇ 5.4 kb to +21 bp of a human aP2 gene (see, e.g., Tozzo et al. (1997) Endocrinol. 138:1604; Ross et al. (1990) Proc. Natl. Acad. Sci. USA 87:9590; and Pavjani et al. (2005) Nat. Med. 11:797); a glucose transporter-4 (GLUT4) promoter (see, e.g., Knight et al. (2003) Proc. Natl. Acad. Sci.
  • aP2 gene promoter/enhancer e.g., a region from ⁇ 5.4 kb to +21 bp of a human aP2 gene (see, e.g., Tozzo et al. (1997) Endocrinol. 138:160
  • fatty acid translocase (FAT/CD36) promoter see, e.g., Kuriki et al. (2002) Biol. Pharm. Bull. 25:1476; and Sato et al. (2002) J. Biol. Chem. 277:15703
  • SCD1 stearoyl-CoA desaturase-1
  • SCD1 stearoyl-CoA desaturase-1 promoter
  • leptin promoter see, e.g., Mason et al. (1998 Endocrinol. 139:1013; and Chen et al. (1999) Biochem. Biophys. Res. Comm.
  • an adiponectin promoter see, e.g., Kita et al. (2005) Biochem. Biophys. Res. Comm. 331:484; and Chakrabarti (2010) Endocrinol. 151:2408
  • an adipsin promoter see, e.g., Platt et al. (1989) Proc. Natl. Acad. Sci. USA 86:7490
  • a resistin promoter see, e.g., Seo et al. (2003) Molec. Endocrinol. 17:1522); and the like.
  • Cardiomyocyte-specific spatially restricted promoters include, but are not limited to control sequences derived from the following genes: myosin light chain-2, ⁇ -myosin heavy chain, AE3, cardiac troponin C, cardiac actin, and the like.
  • Franz et al. (1997) Cardiovasc. Res. 35:560-566; Robbins et al. (1995) Ann. N.Y. Acad. Sci. 752:492-505; Linn et al. (1995) Circ. Res. 76:584-591; Parmacek et al. (1994) Mol. Cell. Biol. 14:1870-1885; Hunter et al. (1993) Hypertension 22:608-617; and Sartorelli et al. (1992) Proc. Natl. Acad. Sci. USA 89:4047-4051.
  • Smooth muscle-specific spatially restricted promoters include, but are not limited to an SM22 ⁇ promoter (see, e.g., Akyürek et al. (2000) Mol. Med. 6:983; and U.S. Pat. No. 7,169,874); a smoothelin promoter (see, e.g., WO 2001/018048; an ⁇ -smooth muscle actin promoter; and the like.
  • a 0.4 kb region of the SM22 ⁇ promoter, within which lie two CArG elements has been shown to mediate vascular smooth muscle cell-specific expression (see, e.g., Kim, et al. (1997) Mol. Cell. Biol. 17, 2266-2278; Li, et al., (1996) J. Cell Biol. 132, 849-859; and Moessler, et al. (1996) Development 122, 2415-2425).
  • Photoreceptor-specific spatially restricted promoters include, but are not limited to, a rhodopsin promoter; a rhodopsin kinase promoter (Young et al. (2003) Ophthalmol. Vis. Sci. 44:4076); a beta phosphodiesterase gene promoter (Nicoud et al. (2007) J. Gene Med. 9:1015); a retinitis pigmentosa gene promoter (Nicoud et al. (2007) supra); an interphotoreceptor retinoid-binding protein (IRBP) gene enhancer (Nicoud et al. (2007) supra); an IRBP gene promoter (Yokoyama et al. (1992) Exp Eye Res. 55:225); and the like.
  • a rhodopsin promoter a rhodopsin kinase promoter
  • a beta phosphodiesterase gene promoter Necoud et al. (2007) J. Gene
  • nucleic acid e.g., an expression construct
  • Suitable methods include e.g., viral or bacteriophage infection, transfection, conjugation, protoplast fusion, lipofection, electroporation, calcium phosphate precipitation, polyethyleneimine (PEI)-mediated transfection, DEAE-dextran mediated transfection, liposome-mediated transfection, particle gun technology, calcium phosphate precipitation, direct micro injection, nanoparticle-mediated nucleic acid delivery (see, e.g., Panyam et., al Adv Drug Deliv Rev. 2012 Sep. 13. pii: 50169-409X(12)00283-9. doi: 10.1016/j.addr.2012.09.023), and the like.
  • PKI polyethyleneimine
  • Contacting cells with a Cas9 targeting complex may occur in any culture media and under any culture conditions that promote the survival of the cells.
  • cells may be suspended in any appropriate nutrient medium that is convenient, such as Iscove's modified DMEM or RPMI 1640, supplemented with fetal calf serum or heat inactivated goat serum (about 5-10%), L-glutamine, a thiol, particularly 2-mercaptoethanol, and antibiotics, e.g. penicillin and streptomycin.
  • the culture may contain growth factors to which the cells are responsive. Growth factors, as defined herein, are molecules capable of promoting survival, growth and/or differentiation of cells, either in culture or in the intact tissue, through specific effects on a transmembrane receptor. Growth factors include polypeptides and non-polypeptide factors. Conditions that promote the survival of cells are typically permissive of the subject cleavage and binding methods
  • a Cas9 protein can be codon optimized.
  • a codon optimized Cas9 protein is a variant Cas9 protein.
  • a codon optimized Cas9 protein is a chimeric Cas9 protein. Codon optimization is known in the art and entails the mutation of foreign-derived DNA to mimic the codon preferences of the intended host organism or host cell while encoding the same protein. Thus, the codons are changed, but the encoded protein remains unchanged. For example, if the intended target cell was a human cell, a human codon optimized Cas9 (or Cas9 variant) would be a suitable Cas9 protein.
  • a mouse codon optimized Cas9 or variant, e.g., enzymatically inactive variant
  • a suitable Cas9 protein While codon optimization is not required, it is acceptable and may be preferable in certain cases.
  • a component of a Cas9 targeting complex e.g., a targeter-RNA, an activator-RNA, a guide RNA (e.g., a single guide RNA), and/or a Cas9 protein
  • the component of a Cas9 targeting complex can be produced by direct chemical synthesis or may be transcribed in vitro from a DNA (e.g., encoding the component). Methods of synthesizing RNA from a DNA template are well known in the art.
  • the component or components will be synthesized in vitro using an RNA polymerase enzyme (e.g., T7 polymerase, T3 polymerase, SP6 polymerase, etc.).
  • the RNA may be introduced into a cell by any of the well-known techniques for introducing nucleic acids into cells (e.g., microinjection, electroporation, transfection, nucleofection, etc).
  • a component of a Cas9 targeting complex can be produced using any convenient method (e.g., chemical synthesis).
  • any of the components of a Cas9 targeting complex may be provided to the cells using any convenient transfection techniques; see, e.g. Angel and Yanik (2010) PLoS ONE 5(7): e11756, and the commercially available TransMessenger® reagents from Qiagen, StemfectTM RNA Transfection Kit from Stemgent, and TransIT®-mRNA Transfection Kit from Mirus Bio LLC. See also Beumer et al. (2008 Efficient gene targeting in Drosophila by direct embryo injection with zinc-finger nucleases. PNAS 105(50):19821-19826.
  • nucleic acids encoding any of the components of a Cas9 targeting complex may be provided on DNA vectors. Many vectors, e.g.
  • the vectors comprising the nucleic acid(s) may be maintained episomally, e.g. as plasmids, minicircle DNAs, viruses such cytomegalovirus, adenovirus, etc., or they may be integrated into the target cell genome, through homologous recombination or random integration, e.g. retrovirus-derived vectors such as MMLV, HIV-1, ALV, etc.
  • a targeting complex can be formed (e.g., in vitro) prior to contact with a target DNA (e.g., prior to introduction into a cell).
  • a Cas9 protein is combined with a guide RNA (thus forming a ribonucleoprotein (RNP) that can be referred to as a targeting complex), prior to introduction into a cell.
  • Delivery of the targeting complex (the resulting RNP) can be achieved by any convenient method (e.g., nucleofection, direct injection, or any other method of delivering nucleic acid and/or protein to a cell, e.g., as discussed herein).
  • Vectors may be provided directly to the subject cells.
  • the cells are contacted with vector(s) comprising the nucleic acid(s) encoding a component(s) of a Cas9 targeting complex such that the vector(s) is/are taken up by the cells.
  • Methods for contacting cells with nucleic acid vectors that are plasmids including electroporation, calcium chloride transfection, microinjection, and lipofection are well known in the art.
  • the cells are contacted with viral particles comprising the nucleic acid encoding at least one component of a Cas9 targeting complex.
  • Retroviruses for example, lentiviruses, are particularly suitable to the method of the invention.
  • retroviral vectors are “defective”, i.e. unable to produce viral proteins required for productive infection. Rather, replication of the vector requires growth in a packaging cell line.
  • the retroviral nucleic acids comprising the nucleic acid are packaged into viral capsids by a packaging cell line.
  • Different packaging cell lines provide a different envelope protein (ecotropic, amphotropic or xenotropic) to be incorporated into the capsid, this envelope protein determining the specificity of the viral particle for the cells (ecotropic for murine and rat; amphotropic for most mammalian cell types including human, dog and mouse; and xenotropic for most mammalian cell types except murine cells).
  • the appropriate packaging cell line may be used to ensure that the cells are targeted by the packaged viral particles.
  • Methods of introducing the retroviral vectors comprising the nucleic acid encoding the reprogramming factors into packaging cell lines and of collecting the viral particles that are generated by the packaging lines are well known in the art.
  • Nucleic acids can also introduced by direct micro-injection (e.g., injection of RNA into a zebrafish embryo).
  • Vectors used for providing the nucleic acids encoding a component(s) of a Cas9 targeting complex to the subject cells will typically comprise suitable promoters for driving the expression, that is, transcriptional activation, of the nucleic acid of interest.
  • the nucleic acid of interest will be operably linked to a promoter (e.g., a eukaryotic promoter).
  • a promoter e.g., a eukaryotic promoter.
  • This may include ubiquitously acting promoters, for example, the CMV- ⁇ -actin promoter, or inducible promoters, such as promoters that are active in particular cell populations or that respond to the presence of drugs such as tetracycline.
  • vectors used for providing a component of a Cas9 targeting complex may include nucleic acid sequences that encode for selectable markers in the target cells, so as to identify cells that have taken up the vectors.
  • a subject component of a Cas9 targeting complex may be used to contact DNA or may be introduced into cells as RNA.
  • Methods of introducing RNA into cells are known in the art and may include, for example, direct injection, transfection, or any other method used for the introduction of DNA.
  • a subject Cas9 protein may be provided to cells as a polypeptide.
  • a polypeptide may optionally be fused to a polypeptide domain (e.g., to increase solubility of the product, to allow for affinity purification, etc.).
  • the domain may be linked to the polypeptide through a defined protease cleavage site, e.g. a TEV sequence, which is cleaved by TEV protease.
  • the linker may also include one or more flexible sequences, e.g. from 1 to 10 glycine residues.
  • the cleavage of the fusion protein is performed in a buffer that maintains solubility of the product, e.g.
  • Domains of interest include endosomolytic domains, e.g. influenza HA domain; and other polypeptides that aid in production, e.g. IF2 domain, GST domain, MBP domain, His tag, HA tag, FLAG tag, GRPE domain, and the like.
  • the polypeptide may be formulated for improved stability.
  • the peptides may be PEGylated, where the polyethyleneoxy group provides for enhanced lifetime in the blood stream.
  • the subject Cas9 protein may be fused to a polypeptide permeant domain to promote uptake by the cell.
  • a polypeptide permeant domain may be fused to a polypeptide permeant domain to promote uptake by the cell.
  • permeant domains are known in the art and may be used in the non-integrating polypeptides of the present invention, including peptides, peptidomimetics, and non-peptide carriers.
  • a permeant peptide may be derived from the third alpha helix of Drosophila melanogaster transcription factor Antennapaedia, referred to as penetratin, which comprises the amino acid sequence RQIKIWFQNRRMKWKK (SEQ ID NO: 268)
  • the permeant peptide comprises the HIV-1 tat basic region amino acid sequence, which may include, for example, amino acids 49-57 of naturally-occurring tat protein.
  • Other permeant domains include polyarginine motifs, for example, the region of amino acids 34-56 of HIV-1 rev protein, nona-arginine, octa-arginine, and the like. (See, for example, Futaki et al.
  • the nona-arginine (R9) sequence is one of the more efficient PTDs that have been characterized (Wender et al. 2000; Uemura et al. 2002).
  • the site at which the fusion is made may be selected in order to optimize the biological activity, secretion or binding characteristics of the polypeptide. The optimal site will be determined by routine experimentation.
  • a subject Cas9 protein may be produced in vitro or by eukaryotic cells or by prokaryotic cells, and it may be further processed by unfolding, e.g. heat denaturation, DTT reduction, etc. and may be further refolded, using methods known in the art.
  • Modifications of interest that do not alter primary sequence include chemical derivatization of polypeptides, e.g., acylation, acetylation, carboxylation, amidation, etc. Also included are modifications of glycosylation, e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
  • modifications of glycosylation e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or
  • targeter-RNAs include those containing residues other than naturally occurring L-amino acids, e.g. D-amino acids or non-naturally occurring synthetic amino acids. D-amino acids may be substituted for some or all of the amino acid residues.
  • the Cas9 proteins may be prepared by in vitro synthesis, using conventional methods as known in the art.
  • Various commercial synthetic apparatuses are available, for example, automated synthesizers by Applied Biosystems, Inc., Beckman, etc. By using synthesizers, naturally occurring amino acids may be substituted with unnatural amino acids. The particular sequence and the manner of preparation will be determined by convenience, economics, purity required, and the like.
  • cysteines can be used to make thioethers, histidines for linking to a metal ion complex, carboxyl groups for forming amides or esters, amino groups for forming amides, and the like.
  • the Cas9 proteins may also be isolated and purified in accordance with conventional methods of recombinant synthesis (e.g., using an affinity tag such as a HIS tag, and HA tag, etc.).
  • a lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique.
  • the compositions which are used will comprise 20% or more by weight of the desired product, more usually 75% or more by weight, preferably 95% or more by weight, and for therapeutic purposes, usually 99.5% or more by weight, in relation to contaminants related to the method of preparation of the product and its purification. Usually, the percentages will be based upon total protein.
  • the guide RNA and/or the Cas9 protein are provided to the cells for about 30 minutes to about 24 hours, e.g., 1 hour, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 12 hours, 16 hours, 18 hours, 20 hours, or any other period from about 30 minutes to about 24 hours, which may be repeated with a frequency of about every day to about every 4 days, e.g., every 1.5 days, every 2 days, every 3 days, or any other frequency from about every day to about every four days.
  • the agent(s) may be provided to the subject cells one or more times, e.g. one time, twice, three times, or more than three times, and the cells allowed to incubate with the agent(s) for some amount of time following each contacting event e.g. 16-24 hours, after which time the media is replaced with fresh media and the cells are cultured further.
  • the complexes may be provided simultaneously (e.g. as two polypeptides and/or nucleic acids), or delivered simultaneously. Alternatively, they may be provided consecutively, e.g. the targeting complex being provided first, followed by the second targeting complex, etc. or vice versa.
  • a subject nucleic acid e.g., a guide RNA, a PAM-mer, etc.
  • a nucleoside is a base-sugar combination.
  • the base portion of the nucleoside is normally a heterocyclic base.
  • the two most common classes of such heterocyclic bases are the purines and the pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate group can be linked to the 2′, the 3′, or the 5′ hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • the respective ends of this linear polymeric compound can be further joined to form a circular compound, however, linear compounds are suitable.
  • linear compounds may have internal nucleotide base complementarity and may therefore fold in a manner as to produce a fully or partially double-stranded compound.
  • the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide.
  • the normal linkage or backbone of RNA and DNA is a 3′ to 5′ phosphodiester linkage.
  • Suitable nucleic acid modifications include, but are not limited to: 2′Omethyl modified nucleotides, 2′ Fluoro modified nucleotides, locked nucleic acid (LNA) modified nucleotides, peptide nucleic acid (PNA) modified nucleotides, nucleotides with phosphorothioate linkages, and a 5′ cap (e.g., a 7-methylguanylate cap (m7G)). Additional details and additional modifications are described below.
  • LNA locked nucleic acid
  • PNA peptide nucleic acid
  • a 2′-O-Methyl modified nucleotide (also referred to as 2′-O-Methyl RNA) is a naturally occurring modification of RNA found in tRNA and other small RNAs that arises as a post-transcriptional modification. Oligonucleotides can be directly synthesized that contain 2′-O-Methyl RNA. This modification increases Tm of RNA:RNA duplexes but results in only small changes in RNA:DNA stability. It is stabile with respect to attack by single-stranded ribonucleases and is typically 5 to 10-fold less susceptible to DNases than DNA. It is commonly used in antisense oligos as a means to increase stability and binding affinity to the target message.
  • 2′ Fluoro modified nucleotides e.g., 2′ Fluoro bases
  • 2′ Fluoro bases have a fluorine modified ribose which increases binding affinity (Tm) and also confers some relative nuclease resistance when compared to native RNA.
  • Tm binding affinity
  • siRNAs are commonly employed in ribozymes and siRNAs to improve stability in serum or other biological fluids.
  • LNA bases have a modification to the ribose backbone that locks the base in the C3′-endo position, which favors RNA A-type helix duplex geometry. This modification significantly increases Tm and is also very nuclease resistant. Multiple LNA insertions can be placed in an oligo at any position except the 3′-end. Applications have been described ranging from antisense oligos to hybridization probes to SNP detection and allele specific PCR. Due to the large increase in Tm conferred by LNAs, they also can cause an increase in primer dimer formation as well as self-hairpin formation. In some cases, the number of LNAs incorporated into a single oligo is 10 bases or less.
  • the phosphorothioate (PS) bond (i.e., a phosphorothioate linkage) substitutes a sulfur atom for a non-bridging oxygen in the phosphate backbone of a nucleic acid (e.g., an oligo). This modification renders the internucleotide linkage resistant to nuclease degradation.
  • Phosphorothioate bonds can be introduced between the last 3-5 nucleotides at the 5′- or 3′-end of the oligo to inhibit exonuclease degradation. Including phosphorothioate bonds within the oligo (e.g., throughout the entire oligo) can help reduce attack by endonucleases as well.
  • a subject nucleic acid (e.g., a guide RNA, a PAM-mer, etc.) has one or more nucleotides that are 2′-O-Methyl modified nucleotides.
  • a subject nucleic acid (e.g., a guide RNA, a PAM-mer, etc.) has one or more 2′ Fluoro modified nucleotides.
  • a subject nucleic acid e.g., a guide RNA, a PAM-mer, etc.
  • LNA bases e.g., LNA bases.
  • a subject nucleic acid (e.g., a guide RNA, a PAM-mer, etc.) has one or more nucleotides that are linked by a phosphorothioate bond (i.e., the subject nucleic acid has one or more phosphorothioate linkages).
  • a subject nucleic acid e.g., a guide RNA, a PAM-mer, etc.
  • has a 5′ cap e.g., a 7-methylguanylate cap (m7G)
  • a subject nucleic acid (e.g., a guide RNA, a PAM-mer, etc.) has a combination of modified nucleotides.
  • a subject nucleic acid e.g., a guide RNA, a PAM-mer, etc.
  • a 5′ cap e.g., a 7-methylguanylate cap (m7G)
  • m7G 7-methylguanylate cap
  • a 2′-O-Methyl nucleotide and/or a 2′ Fluoro modified nucleotide and/or a LNA base and/or a phosphorothioate linkage e.g., a 2′-O-Methyl nucleotide and/or a 2′ Fluoro modified nucleotide and/or a LNA base and/or a phosphorothioate linkage.
  • a subject guide RNA has one or more nucleotides that are 2′-O-Methyl modified nucleotides. In some embodiments, a subject guide RNA has one or more 2′ Fluoro modified nucleotides. In some embodiments, a subject guide RNA has one or more LNA bases. In some embodiments, a subject guide RNA has one or more nucleotides that are linked by a phosphorothioate bond (i.e., the subject nucleic acid has one or more phosphorothioate linkages). In some embodiments, a subject guide RNA has a 5′ cap (e.g., a 7-methylguanylate cap (m7G)).
  • m7G 7-methylguanylate cap
  • a subject guide RNA has a combination of modified nucleotides.
  • a subject guide RNA can have a 5′ cap (e.g., a 7-methylguanylate cap (m7G)) in addition to having one or more nucleotides with other modifications (e.g., a 2′-O-Methyl nucleotide and/or a 2′ Fluoro modified nucleotide and/or a LNA base and/or a phosphorothioate linkage).
  • m7G 7-methylguanylate cap
  • nucleic acids containing modifications include nucleic acids containing modified backbones or non-natural internucleoside linkages.
  • Nucleic acids having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • Suitable modified oligonucleotide backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, phosphorodiamidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′
  • Suitable oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e. a single inverted nucleoside residue which may be a basic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts such as, for example, potassium or sodium), mixed salts and free acid forms are also included.
  • a subject nucleic acid comprises one or more phosphorothioate and/or heteroatom internucleoside linkages, in particular —CH 2 —NH—O—CH 2 —, —CH 2 —N(CH 3 )—O—CH 2 — (known as a methylene (methylimino) or MMI backbone), —CH 2 —O—N(CH 3 )—CH 2 —, —CH 2 —N(CH 3 )—N(CH 3 )—CH 2 — and —O—N(CH 3 )—CH 2 —CH 2 — (wherein the native phosphodiester internucleotide linkage is represented as —O—P( ⁇ O)(OH)—O—CH 2 —).
  • MMI type internucleoside linkages are disclosed in the above referenced U.S. Pat. No. 5,489,677. Suitable amide internucleoside linkages are disclosed in t U.S. Pat. No. 5,602,
  • nucleic acids having morpholino backbone structures as described in, e.g., U.S. Pat. No. 5,034,506.
  • a subject nucleic acid comprises a 6-membered morpholino ring in place of a ribose ring.
  • a phosphorodiamidate or other non-phosphodiester internucleoside linkage replaces a phosphodiester linkage.
  • Suitable modified polynucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • riboacetyl backbones alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • a subject nucleic acid can be a nucleic acid mimetic.
  • the term “mimetic” as it is applied to polynucleotides is intended to include polynucleotides wherein only the furanose ring or both the furanose ring and the internucleotide linkage are replaced with non-furanose groups, replacement of only the furanose ring is also referred to in the art as being a sugar surrogate.
  • the heterocyclic base moiety or a modified heterocyclic base moiety is maintained for hybridization with an appropriate target DNA.
  • One such nucleic acid, a polynucleotide mimetic that has been shown to have excellent hybridization properties is referred to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-backbone of a polynucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • the nucleotides are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • PNA peptide nucleic acid
  • the backbone in PNA compounds is two or more linked aminoethylglycine units which gives PNA an amide containing backbone.
  • the heterocyclic base moieties are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative U.S. patents that describe the preparation of PNA compounds include, but are not limited to: U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262.
  • Another class of polynucleotide mimetic that has been studied is based on linked morpholino units (morpholino nucleic acid) having heterocyclic bases attached to the morpholino ring.
  • a number of linking groups have been reported that link the morpholino monomeric units in a morpholino nucleic acid.
  • One class of linking groups has been selected to give a non-ionic oligomeric compound.
  • the non-ionic morpholino-based oligomeric compounds are less likely to have undesired interactions with cellular proteins.
  • Morpholino-based polynucleotides are non-ionic mimics of oligonucleotides which are less likely to form undesired interactions with cellular proteins (Dwaine A. Braasch and David R.
  • Morpholino-based polynucleotides are disclosed in U.S. Pat. No. 5,034,506. A variety of compounds within the morpholino class of polynucleotides have been prepared, having a variety of different linking groups joining the monomeric subunits.
  • CeNA cyclohexenyl nucleic acids
  • the furanose ring normally present in a DNA/RNA molecule is replaced with a cyclohexenyl ring.
  • CeNA DMT protected phosphoramidite monomers have been prepared and used for oligomeric compound synthesis following classical phosphoramidite chemistry.
  • Fully modified CeNA oligomeric compounds and oligonucleotides having specific positions modified with CeNA have been prepared and studied (see Wang et al., J. Am. Chem. Soc., 2000, 122, 8595-8602). In general the incorporation of CeNA monomers into a DNA chain increases its stability of a DNA/RNA hybrid.
  • CeNA oligoadenylates formed complexes with RNA and DNA complements with similar stability to the native complexes.
  • the study of incorporating CeNA structures into natural nucleic acid structures was shown by NMR and circular dichroism to proceed with easy conformational adaptation.
  • a further modification includes Locked Nucleic Acids (LNAs) in which the 2′-hydroxyl group is linked to the 4′ carbon atom of the sugar ring thereby forming a 2′-C,4′-C-oxymethylene linkage thereby forming a bicyclic sugar moiety.
  • the linkage can be a methylene (—CH 2 —), group bridging the 2′ oxygen atom and the 4′ carbon atom wherein n is 1 or 2 (Singh et al., Chem. Commun., 1998, 4, 455-456).
  • Potent and nontoxic antisense oligonucleotides containing LNAs have been described (e.g., Wahlestedt et al., Proc. Natl. Acad. Sci. U.S.A., 2000, 97, 5633-5638.
  • LNAs and preparation thereof are also described in WO 98/39352 and WO 99/14226, as well as U.S. applications 20120165514, 20100216983, 20090041809, 20060117410, 20040014959, 20020094555, and 20020086998.
  • a subject nucleic acid can also include one or more substituted sugar moieties.
  • Suitable polynucleotides comprise a sugar substituent group selected from: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C.sub.1 to C 10 alkyl or C 2 to C 10 alkenyl and alkynyl.
  • Suitable polynucleotides comprise a sugar substituent group selected from: C 1 to C 10 lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a sugar substituent group selected from: C 1 to C 10 lower alkyl,
  • a suitable modification includes 2′-methoxyethoxy (2′-O—CH 2 CH 2 OCH 3 , also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group.
  • a further suitable modification includes 2′-dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2′-DMAOE, as described in examples hereinbelow, and 2′-dimethylaminoethoxyethoxy (also known in the art as 2′-O-dimethyl-amino-ethoxy-ethyl or 2′-DMAEOE), i.e., 2′-O—CH 2 —O—CH 2 —N(CH 3 ) 2 .
  • sugar substituent groups include methoxy (—O—CH 3 ), aminopropoxy (—O CH 2 CH 2 CH 2 NH 2 ), allyl (—CH 2 —CH ⁇ CH 2 ), —O-allyl (—O—CH 2 —CH ⁇ CH 2 ) and fluoro (F).
  • 2′-sugar substituent groups may be in the arabino (up) position or ribo (down) position.
  • a suitable 2′-arabino modification is 2′-F.
  • Similar modifications may also be made at other positions on the oligomeric compound, particularly the 3′ position of the sugar on the 3′ terminal nucleoside or in 2′-5′ linked oligonucleotides and the 5′ position of 5′ terminal nucleotide.
  • Oligomeric compounds may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
  • a subject nucleic acid may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C ⁇ C—CH 3 ) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and gu
  • nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(1H-pyrimido(5,4-b)(1,4)benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido(5,4-b)(1,4)benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g.
  • Heterocyclic base moieties may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone.
  • Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y.
  • nucleobases are useful for increasing the binding affinity of an oligomeric compound.
  • These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C.
  • Another possible modification of a subject nucleic acid involves chemically linking to the polynucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties or conjugates can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups.
  • Conjugate groups include, but are not limited to, intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers.
  • Suitable conjugate groups include, but are not limited to, cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Groups that enhance the pharmacodynamic properties include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence-specific hybridization with the target nucleic acid.
  • Groups that enhance the pharmacokinetic properties include groups that improve uptake, distribution, metabolism or excretion of a subject nucleic acid.
  • Conjugate moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem.
  • lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053
  • Acids Res., 1990, 18, 3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937.
  • a conjugate may include a “Protein Transduction Domain” or PTD (also known as a CPP—cell penetrating peptide), which may refer to a polypeptide, polynucleotide, carbohydrate, or organic or inorganic compound that facilitates traversing a lipid bilayer, micelle, cell membrane, organelle membrane, or vesicle membrane.
  • PTD Protein Transduction Domain
  • a PTD attached to another molecule which can range from a small polar molecule to a large macromolecule and/or a nanoparticle, facilitates the molecule traversing a membrane, for example going from extracellular space to intracellular space, or cytosol to within an organelle.
  • a PTD is covalently linked to the amino terminus of an exogenous polypeptide (e.g., a Cas9 protein). In some embodiments, a PTD is covalently linked to the carboxyl terminus of an exogenous polypeptide (e.g., a Cas9 protein). In some embodiments, a PTD is covalently linked to a nucleic acid (e.g., a guide RNA, a polynucleotide encoding a guide RNA, a polynucleotide encoding a Cas9 protein, etc.).
  • a nucleic acid e.g., a guide RNA, a polynucleotide encoding a guide RNA, a polynucleotide encoding a Cas9 protein, etc.
  • Exemplary PTDs include but are not limited to a minimal undecapeptide protein transduction domain (corresponding to residues 47-57 of HIV-1 TAT comprising YGRKKRRQRRR; SEQ ID NO:264); a polyarginine sequence comprising a number of arginines sufficient to direct entry into a cell (e.g., 3, 4, 5, 6, 7, 8, 9, 10, or 10-50 arginines); a VP22 domain (Zender et al. (2002) Cancer Gene Ther. 9(6):489-96); an Drosophila Antennapedia protein transduction domain (Noguchi et al. (2003) Diabetes 52(7):1732-1737); a truncated human calcitonin peptide (Trehin et al. (2004) Pharm.
  • a minimal undecapeptide protein transduction domain corresponding to residues 47-57 of HIV-1 TAT comprising YGRKKRRQRRR; SEQ ID NO:264
  • a polyarginine sequence comprising a number of arginines
  • Exemplary PTDs include but are not limited to, YGRKKRRQRRR (SEQ ID NO:264), RKKRRQRRR (SEQ ID NO:269); an arginine homopolymer of from 3 arginine residues to 50 arginine residues;
  • Exemplary PTD domain amino acid sequences include, but are not limited to, any of the following: YGRKKRRQRRR (SEQ ID NO:264); RKKRRQRR (SEQ ID NO:270); YARAAARQARA (SEQ ID NO:271); THRLPRRRRRR (SEQ ID NO:272); and GGRRARRRRRR (SEQ ID NO:273).
  • the PTD is an activatable CPP (ACPP) (Aguilera et al. (2009) Integr Biol ( Camb ) June; 1(5-6): 371-381).
  • ACPPs comprise a polycationic CPP (e.g., Arg9 or “R9”) connected via a cleavable linker to a matching polyanion (e.g., Glu9 or “E9”), which reduces the net charge to nearly zero and thereby inhibits adhesion and uptake into cells.
  • a polyanion e.g., Glu9 or “E9”
  • kits for carrying out a subject method can include a cell cycle blocking agent and one or more of: (i) a single guide RNA, (ii) a DNA polynucleotide encoding a single guide RNA, (iii) a targeter-RNA, (iv) a DNA polynucleotide encoding a targeter-RNA, (v) an activator-RNA, (vi) a DNA polynucleotide encoding an activator-RNA, (vii) a Cas9 protein, and (h) a nucleic acid encoding a Cas9 protein; all of which are described in detail above.
  • a kit includes a donor polynucleotide (e.g, encoding a marker protein, e.g., a fluorescent protein such as GFP, RFP, CFP, YFP, and the like).
  • a marker protein e.g., a fluorescent protein such as GFP, RFP, CFP, YFP, and the like.
  • Cas9 protein encompasses a variant Cas9 protein, a wild type Cas9 protein, a chimeric Cas9 protein, etc.
  • kits can further include one or more additional reagents, where such additional reagents can be selected from: a dilution buffer; a reconstitution solution; a wash buffer; a control reagent; a control expression vector or RNA polynucleotide; a reagent for in vitro production of the Cas9 protein from DNA, and the like.
  • additional reagents can be selected from: a dilution buffer; a reconstitution solution; a wash buffer; a control reagent; a control expression vector or RNA polynucleotide; a reagent for in vitro production of the Cas9 protein from DNA, and the like.
  • a subject kit comprises a variant Cas9 protein (or a nucleotide encoding the same) that exhibits reduced nuclease activity relative to wild-type Cas9.
  • Components of a subject kit can be in separate containers; or can be combined in a single container.
  • a subject kit can further include instructions for using the components of the kit to practice the subject methods.
  • the instructions for practicing the subject methods are generally recorded on a suitable recording medium.
  • the instructions may be printed on a substrate, such as paper or plastic, etc.
  • the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or subpackaging) etc.
  • the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g. CD-ROM, diskette, flash drive, etc.
  • the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g. via the internet, are provided.
  • An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, this means for obtaining the instructions is recorded on a suitable substrate.
  • Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pl, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal(ly); s.c., subcutaneous(ly); and the like.
  • the CRISPR/Cas9 system is a robust genome editing technology that functions in human cells, animals and plants based on the RNA-programmed DNA cleaving activity of the Cas9 enzyme.
  • Homology-directed repair (HDR) of Cas9-induced site-specific double-strand DNA breaks leads to integration of donor DNA sequences at the break site, enabling targeted genome engineering.
  • the more prevalent repair pathway non-homologous end joining (NHEJ) generates small random insertions or deletions (indels) at the site of the original break.
  • NHEJ non-homologous end joining
  • indels small random insertions or deletions
  • Cas9 RNP-mediated HDR in HEK293T primary human fibroblasts and human embryonic stem cells was increased by several fold relative to experiments in unsynchronized cells, while cell mortality and off-target effects were minimized. This approach provides a simple and highly effective strategy for enhancing site-specific genome engineering in both transformed and primary cells.
  • DMEM media fetal bovine serum, non-essential amino acid, penicillin-streptomycin, DPBS and 0.05% trypsin were purchased from Life technologies.
  • 293T cells were maintained in DMEM media supplemented with 10% fetal bovine serum, non-essential amino acid and penicillin-streptomycin.
  • Human neonatal Fibroblasts were maintained in 15% fetal bovine serum, non-essential amino acid and penicillin-streptomycin.
  • H9 human embryonic stem cells were maintained on Matrigel in E8 media plus supplement.
  • Aphidicolin, hydroxyurea, lovastatin, mimosine, nocodazole and thymidine were purchased from Sigma-Aldrich.
  • the synchronization protocols were modified from the following references (Adams et al., 1967, J. Biol. Chem. 242, 1314-1317; Harper et al., 2007, Methods in Molecular Biology (Clifton, N.J.) 296, 157-166; Jackman et al., 2001, Curr Protoc Cell Biol Chapter 8, Unit8.3; Pauklin et al., 2003, Cell 155, 135-147). It is important to ensure cells are maintained at ⁇ 70% confluency.
  • HEK293T cells were seeded at 1 ⁇ 10 6 cell density in a 10-cm culture dish. Primary neonatal fibroblast cells were seeded at 5 ⁇ 10 5 in 10-cm dish. Human ES cells were maintained in 6 well dishes. Aphidicolin and thymidine require two sequential treatments to enrich cells arrested at the entry of S phase. Cells were treated with aphidicolin (2 ⁇ g/ml) or thymidine (5 mM) for 17 h, washed with media to remove the drugs, grown for 8 h, and treated with a second dose of drugs for 17 h.
  • Hydroxyurea (2 mM), lovastatin (40 ⁇ M), mimosine (200 ⁇ M) and nocodazole (200 ng/ml) require only one treatment for 17 h.
  • Two synchronization conditions were tested in the human ES cell experiment. The first condition was a simple nocodazole treatment for 16 h. The second condition was modified from Pauklin et al., 2003, Cell 155, 135-147. The cells were treated with nocodazole for 16 h, washed to remove the drug, and then treated with aphidicolin for 3 h before nucleofection.
  • the recombinant S. pyogenes Cas9 used in this study carries at C-terminus an HA tag and two nuclear localization signal peptides which facilitates transport across nuclear membrane.
  • the protein was expressed with a N-terminal hexahistidine tag and maltose binding protein in E. coli Rosetta 2 cells (EMD Millipore) from plasmid pMJ915.
  • E. coli Rosetta 2 cells E. coli Rosetta 2 cells (EMD Millipore) from plasmid pMJ915.
  • the His tag and maltose binding protein were cleaved by TEV protease, and Cas9 was purified by the protocols described in Jinek et al 2012 (Jinek et al., 2012, Science 337, 816-821).
  • Cas9 was stored in 20 mM 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid (HEPES) at pH 7.5, 150 mM KCl, 10% glycerol, 1 mM tris(2-chloroethyl) phosphate (TCEP) at ⁇ 80° C.
  • HEP 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid
  • TCEP tris(2-chloroethyl) phosphate
  • the DNA template encoding for a T7 promoter, a 20-nt target sequence and the sgRNA scaffold was assembled from synthetic oligonucleotides by overlapping PCR.
  • the PCR reaction contains 20 nM premix of BS16 (5′-TAA TAC GAC TCA CTA TAG GTC ACC TCC AAT GAC TAG GGG TTT AAG AGC TAT GCT GGA AAC AGC ATA GCA AGT TTA AAT AAG G-3′)(SEQ ID NO:1392) and BS6 (5′-AAA AAA AGC ACC GAC TCG GTG CCA CTT TTT CAA GTT GAT AAC GGA CTA GCC TTA TTT AAA CTT GCT ATG CTG TTT CCA GC-3′) (SEQ ID NO:1393), 1 ⁇ M premix of T25 (5′-TAA TAC GAC TCA CTA TAG-3′) (SEQ ID NO:1394) and BS
  • thermocycler setting consisted of 30 cycles of 95° C. for 10 sec, 57° C. for 10 sec and 72° C. for 10 sec.
  • the PCR product was extracted once with phenol:chloroform:isoamylalcohol and then once with chloroform, before isopropanol precipitation overnight at ⁇ 20° C.
  • the DNA pellet was washed three times with 70% ethanol, dried by vacuum and dissolved in DEPC-treated water.
  • the DYRK1 sgRNA template was assembled from T25, BS6, BS7 and BS14 (5′-TAA TAC GAC TCA CTA TAG GTT CCT TAA ATA AGA ACT TTG TTT AAG AGC TAT GCT GGA AAC AGC ATA GCA AGT TTA AAT AAG G-3′) (SEQ ID NO:1396).
  • a 100- ⁇ 1 T7 in vitro transcription reaction consisted of 30 mM Tris-HCl (pH 8), 20 mM MgCl 2 , 0.01% Triton X-100, 2 mM spermidine, 10 mM fresh dithiothreitol, 5 mM of each ribonucleotide triphosphate, 100 ⁇ g/ml T7 Pol and 1 ⁇ M DNA template.
  • the reaction was incubated at 37° C. for 4 h, and 5 units of RNase-free DNaseI (Promega) was added to digest the DNA template 37° C. for 1 h.
  • RNA was purified by electrophoresis in 10% polyacrylamide gel containing 6M urea.
  • the RNA band was excised from the gel, grinded up in a 15-ml tube, and eluted with 5 volumes of 300 mM sodium acetate (pH 5) overnight at 4° C.
  • One equivalent of isopropanol was added to precipitate the RNA at ⁇ 20° C.
  • the RNA pellet was collected by centrifugation, washed three times with 70% ethanol, and dried by vacuum.
  • the RNA pellet was first dissolved in 20 mM HEPES (pH 7.5), 150 mM KCl, 10% glycerol and 1 mM TCEP. The sgRNA was heated to 70° C. for 5 min and cooled to room temperature. MgCl 2 was added to a final concentration of 1 mM. The sgRNA was again heated to 50° C. for 5 min and cooled to room temperature. Nanodrop and adjust concentration to 100 uM using 20 mM HEPES (pH 7.5), 150 mM KCl, 10% glycerol, 1 mM TCEP, 1 mM MgCl 2 .
  • Double-stranded HDR template 6 and 7 were prepared by PCR amplification.
  • Template 6 was PCR amplified from single-stranded template 5 (5′-TGG CCA GGG AGT GGC CAG AGT CCA GCT TGG GCC CAC GCA GGG GCC TGG CCA GCA GCA AGC AGC ACT CTG CCC TCG TGG GTT TGT GGT TGC GGA TCC AAG CTT TTG GAG GTG ACA TCG ATG TCC TCC CCA TTG GCC TGC TTC GTG GCA ATG CGC CAC CGG TTG ATG TGA TGG GAG CCC TTC TTC TTC TGC TCG-3′ (SEQ ID NO:1397) using primer set (forward 5′-CGA GCA GAA GAA GAA GGG CTC CCA TC-3′ (SEQ ID NO:1398) and reverse 5′-TGG CCA GGG AGT GGC CAG AGT CC-3′) (SEQ ID NO:1399).
  • the PCR reaction was performed using Phusion Polymerase according to manufacturer's protocol (NEB).
  • the thermocycler setting consisted of 30 cycles of 95° C. for 20 sec, 67° C. for 10 sec and 72° C. for 20 sec.
  • the PCR product was extracted once with phenol:chloroform:isoamylalcohol and then once with chloroform, before isopropanol precipitation overnight at ⁇ 20° C.
  • the DNA pellet was washed three times with 70% ethanol, dried by vacuum and dissolved in water. The concentration was determined by Nanodrop (Thermo Scientific).
  • Template 7 was assembled from two fragments (A and B) by overlapping PCR.
  • Fragment A was PCR amplified from HEK293T genomic DNA using the primer set (forward 5′-GCT CAG CCT GAG TGT TGA GGC CCC AGT GGC TGC TCT GG-3′ (SEQ ID NO:1400) and reverse 5′-GTG GTT GCG GAT CCA AGC TTT TGG AGG TGA CAT CGA TGT CCT CCC CAT TGG C-3′) (SEQ ID NO:1401).
  • Fragment B was amplified using the primer set (forward 5′-CAC CTC CAA AAG CTT GGA TCC GCA ACC ACA AAC CCA CGA GGG CAG AGT GCT GCT TGC-3′ (SEQ ID NO:1402) and reverse 5′-TGC GGT GGC GGG CGG GCC CGC CCA GGC AGG CAG GC-3′) (SEQ ID NO:1403). Both reaction were performed using Kapa Hot start high-fidelity polymerase (Kapa Biosystems) in high GC buffer according to the manufacturer's protocol.
  • the thermocycler setting consisted of one cycle of 95° C. for 5 min, 30 cycles of 98° C. for 20 sec, 67° C. for 10 sec and 72° C. for 20 sec, and one cycle of 72° C. for 1 min.
  • Cas9 RNP was prepared immediately before experiment by incubating with sgRNA at 1:1.2 molar ratio in 20 ⁇ M HEPES (pH 7.5), 150 mM KCl, 1 mM MgCl 2 , 10% glycerol and 1 mM TCEP at 37° C. for 10 min. HDR donor DNA was then added to the RNP mixture. Cells were dissociated by 0.05% trypsin, spun down by centrifugation at 400 g for 3 min, and washed once with DPBS.
  • Nucleofection of HEK293T cells was performed using Lonza SF cell-kits and program CM130 in an Amaxa 96-well Shuttle system; for primary neonatal fibroblast, P3 primary cell-line kit and program CA137.
  • Each nucleofection reaction consisted of approximately 2 ⁇ 10 5 cells in 20 ⁇ l of nucleofection reagent and mixed with 10 ⁇ l of RNP:DNA.
  • 100 ⁇ l of DMEM media was added to the well to transfer the cells to tissue culture plates.
  • the cells were incubated at 37° C. for 24 h, the media was removed by aspiration, and 100 ⁇ l of Quick Extraction solution (Epicenter) was added to lyse the cells and extract the genomic DNA.
  • the cell lysate was incubated at 65° C. for 20 min and then 95° C. for 20 min, and stored at ⁇ 20° C.
  • the concentration of genomic DNA was determined by NanoDrop (Thermo Scientific).
  • EMX1 forward 5′-GCC ATC CCC TTC TGT GAA TGT TAG AC-3′ (SEQ ID NO:1404) and 5′-GGA GAT TGG AGA CAC GGA GAG CAG-3′(SEQ ID NO:1405).
  • DYRK1 forward 5′-GAG GAG CTG GTC TGT TGG AGA AGT C-3′(SEQ ID NO:1406) and reverse 5′-CCC AAT CCA TAA TCC CAC GTT GCA TG-3′(SEQ ID NO:1407).
  • the PCR reaction was performed using 200 ng of genomic DNA and Kapa Hot start high-fidelity polymerase (Kapa Biosystems) in high GC buffer according to the manufacturer's protocol.
  • the thermocycler setting consisted of one cycle of 95° C. for 5 min, 30 cycles of 98° C. for 20 sec, 62° C. for 15 sec and 72° C. for 1 min, and one cycle of 72° C. for 1 min.
  • the PCR products were analyzed on 2% agarose gel containing SYBR Safe (Life Technologies). The concentration of PCR DNA was quantitated based on the band intensity relative to a DNA standard using the software Image Lab (Bio-Rad). About 200 ng of PCR DNA was used for T7 endonuclease I and HindIII analyses.
  • T7 endonuclease I assay T7 endonuclease I recognizes and cleaves mismatched heteroduplex DNA which arises from hybridization of wild-type and mutant DNA strands.
  • the hybridization reaction contained 200 ng of PCR DNA in KAPA high GC buffer and 50 mM KCl, and was performed on a thermocycler with the following setting: 95° C., 10 min, 95-85° C. at ⁇ 2° C./sec, 85° C. for 1 min, 85-75° C. at ⁇ 2° C./sec, 75° C. for 1 min, 75-65° C. at ⁇ 2° C./sec, 65° C. for 1 min, 65-55° C.
  • Buffer 2 and 5 units of T7 endonuclease I were added to digest the re-annealed DNA.
  • HindIII directly cleaves PCR DNA containing the newly integrated HindIII restriction sequence as the result of successful HDR.
  • the reaction consisted of 200 ng of PCR DNA and 10 units of HindIII High Fidelity in CutSmart Buffer (NEB). After 2 h of incubation at 37° C., the reaction was quenched with one volume of gel loading dye at 70° C. for 10 min. The product was resolved on 2% agarose gel containing SYBR gold (Life technologies). The band intensity was quantitated using Image Lab. The percentage of HDR was calculated using the following equation (b+c/a+b+c) ⁇ 100, where “a” is the band intensity of DNA substrate and “b” and “c” are the cleavage products.
  • the first objective was to test whether S phase was ideal for HDR in HEK293T cells.
  • the approach was to chemically and reversibly synchronize the cells at G1, S and M phases ( FIG. 1A ).
  • Lovastatin blocks at early G1 and partially at G2/M phase, by inhibiting HMG-CoA reductase and resulting in the depletion of mevalonate, which is an essential substrate for cholesterol synthesis.
  • Mimosine arrests cells at the G1-S border prior to onset of DNA replication by an unknown mechanism.
  • Aphidicolin reversibly inhibits replicative DNA polymerases. Excess thymidine causes feedback inhibition of deoxycytosine synthesis.
  • Hydroxyurea blocks cells at S phase by decreasing the production of deoxyribonucleotide via inhibition of the enzyme ribonucleotide reductase.
  • Nocodazole inhibits microtubule polymerization which is a signature structural feature as cells enter mitosis, causing arrest at G2/M phase.
  • the drugs effects were confirmed by fluorescence activated cell sorter (FACS) analysis of the cell cycle ( FIG. 1B ). FACS analysis of the DNA content in the nocodazole-treated cells indicates a two-fold enrichment of the cells arrested at M phase.
  • FACS fluorescence activated cell sorter
  • HEK293T cells were synchronized and RNP was prepared by loading Cas9 with a single guide RNA (sgRNA) to target exon 3 of the EMX1 gene (Materials and methods), which encodes the EMX1 transcription factor essential for neural development.
  • sgRNA single guide RNA
  • Two doses of RNP were co-nucleofected with a donor DNA which was a linear, 183-nt single-strand DNA oligonucleotide (ssODNA) encoding a HindIII restriction site ( FIG. 3A template 1). Twenty four hours post nucleofection, the transfected HEK293T cells were collected, genomic DNA extracted, and the target region PCR amplified for NHEJ and HDR analyses.
  • the NHEJ frequency a functional readout of Cas9 cleavage activity, was measured by T7 endonuclease I assay and calculated using a formula described by Ran et al (Ran et. al., Cell. 2013 Sep. 12; 154(6):1380-9). Using this formula is necessary, because upon re-annealing, one duplex of mutant DNA can produce two duplexes of mutant:wild-type hybrid, doubling the actual NHEJ frequency.
  • the HDR frequency was determined directly by HindIII digestion, which specifically cleaved the newly integrated HindIII sequence, and calculated from the ratio of DNA product to DNA substrate.
  • lovastatin, mimosine and hydroxyurea greatly decreased the NHEJ and HDR frequencies as compared to the unsynchronized cells ( FIG. 1C and FIG. 1D ).
  • Thymidine slightly reduced the NHEJ and HDR frequencies at 30 ⁇ mol of Cas9 but not at 100 ⁇ mol.
  • Aphidicolin and nocodazole increased the NHEJ frequency slightly at 30 ⁇ mol of Cas9; at 100 ⁇ mol of Cas9, the detection limit by T7 endonuclease I assay has likely reached the maximum.
  • the HDR frequency was increased by aphidicolin and nocodazole.
  • the enhancement was more evident at 30 ⁇ mol of Cas9, 9% in the unsynchronized cells to 15% with aphidicolin and 19% with nocodazole ( FIG. 1D ).
  • the highest HDR frequency was achieved in the triplicate experiment, which was 31% as shown in the representative gel in FIG. 1D , was obtained by nocodazole synchronization at 100 ⁇ mol of Cas9.
  • FIG. 1A-1D depict the effect of cell cycle synchronization on NHEJ and HDR frequencies in HEK293T cells.
  • Lovastatin Lovastatin (Lov) blocks cells in G1 phase by inhibiting cholesterol synthesis.
  • Aphidicolin (Aph), hydroxyurea (Hyd), mimosine (Mim) and thymidine (Thy) disrupt DNA replication, each via a different mechanism, to prevent progression through S phase.
  • Nocodazole (Noc) arrests cells in M phase by inhibiting microtubule polymerization.
  • B FACS analysis reveals the DNA content in the cells that are arrested at different phases of cell cycle.
  • Nocodazole treatment results in the accumulation of 4N DNA as compared to the untreated cells, demonstrating a three-fold enrichment of M-phase arrested cells.
  • C NHEJ
  • HDR HDR
  • FIG. 3A Control reaction (labeled as C) contained 100 ⁇ mol of Cas9 but no sgRNA.
  • NHEJ frequency was estimated by T7 endonuclease I assay using a formula described in Materials and methods; HDR frequency was measure by HindIII digestion.
  • a representative gel with % NHEJ/HDR is shown for each analysis. The mean % NHEJ/HDR and standard deviation (error bars) were calculated from three experiments.
  • the NHEJ frequency was 6-8% at 10 ⁇ mol, 19-27% at 30 ⁇ mol and 46-55% at 100 ⁇ mol Cas9 RNP.
  • the NHEJ frequency was 15-17% at 10 ⁇ mol, 35-37% at 30 ⁇ mol, 46-49% at 100 ⁇ mol Cas9 RNP.
  • Synchronization increased the NHEJ frequency two-fold at 10 ⁇ mol and 1.5-fold at 30 ⁇ mol Cas9 RNP, but the enhancement diminished at 100 ⁇ mol as seen previously.
  • the HDR frequency increased dramatically with synchronization, especially at lower doses of Cas9 RNP.
  • the DYRK1 gene which encodes a dual-specificity tyrosine phosphorylation-regulated kinase important for brain development and Downs Syndrome (Anon et al., 2006, Nature 441, 595-600), was targeted.
  • Two, single-stranded HDR templates of different orientations were assayed: one was complementary (+) to the target sequence and the other was non-complementary ( ⁇ ). Both templates yielded comparable levels of HDR, suggesting that the HDR machinery could use either orientation for repair.
  • the enhancement of HDR by nocodazole synchronization was more dramatic.
  • NHEJ and HDR were barely detectable at 10 picomoles ( ⁇ mol) Cas9 RNP.
  • the frequencies increased to 11-12% NHEJ/2% HDR and 31-32% NHEJ/4% HDR respectively ( FIG. 2B ).
  • the synchronized cells yielded 20-24% NHEJ/5-6% HDR at 10 ⁇ mol, 45-47% NHEJ/12-17% HDR at 30 ⁇ mol and 63-65% NHEJ/24-25% HDR at 100 ⁇ mol Cas9 RNP ( FIG. 2B ).
  • nocodazole synchronization enhanced the NHEJ and HDR frequencies more than two-fold and over six-fold at all doses of Cas9 RNP respectively.
  • the results from targeting of EMX1 and DYRK1 loci demonstrate that nocodazole synchronization is highly effective at enhancing both NHEJ and HDR, especially at low doses of Cas9 RNP.
  • FIG. 2A-2B depict the enhancement of NHEJ and HDR at the EMX1 and DYRK1 loci by nocodazole synchronization.
  • A The effect of nocodazole on the NHEJ and HDR frequencies at EMX1 locus.
  • HEK293T cells were synchronized at M phase with 200 ng/ml of nocodazole for 17 h before nucleofection.
  • three concentrations of Cas9 RNP were assayed in combination with three doses of HDR template (template 1 in FIG. 3A ).
  • the NHEJ frequencies at 10 ⁇ mol of Cas9 RNP in the unsynchronized cells were too low and therefore not determined (ND).
  • HDR template homology arms required for competent homologous recombination was then examined.
  • EMX1 as target, four single-stranded and two double-stranded HDR templates with homology arms ranging from 30 to 250 nt in length were designed ( FIG. 3A ).
  • the HDR templates contained two restriction sites at the center; a HindIII site to the left of cleavage site and a BamHI to the right.
  • Successful recombination of the HDR templates at the target site integrates these sequences, and mutates the NGG PAM and 6 nt of the Cas9 target sequence, preventing Cas9 from re-targeting the recombined locus.
  • the concentration of Cas9 RNP was lowered to 30 ⁇ mol and the HDR template to 50 ⁇ mol.
  • aphidicolin was added immediately after transfection to the growth media to block the cells from entering S phase. We hypothesized this would cause a reduction in HDR frequency due to the inability to enter S phase where HDR pathway is most active.
  • nocodazole synchronization facilitated an overall higher HDR frequency than the unsynchronized cells ( FIG. 3B ).
  • the aphidicolin block significantly reduced the HDR frequency, demonstrating that the cells need to proceed through S phase, and possibly G2 phase as well, to achieve high HDR. It is important to note that complete elimination of HDR events was not observed; this was likely due to some fraction of cells leaking through the aphidicolin block.
  • a single-stranded oligonucleotide with 30-nt homology arms was sufficient for detectable HDR, albeit at low frequency (5%).
  • Extending the length of homology arms to 60 nt increased the HDR frequency by four-fold to 19%.
  • Further extension of the homology arms to 90 nt increased the HDR frequency only slightly to 23% and 20% for complementary (template 4) and non-complementary templates (template 5), respectively. Both complementary and non-complementary orientations were similarly effective as seen before.
  • FIG. 3A-3B depict systematic investigation of DNA templates for efficient HDR at the EMX1 locus.
  • Segment of human EMX1 exon 3 shows the 20-nt target sequence (highlighted in blue), the TGG PAM region (in red) and the Cas9 cleavage site at three bases upstream from PAM. Seven HDR templates were tested for HDR efficiency.
  • Template 1 is 183-nt long and contains an 9-nt insertion upstream of the cut site, an 9-nt deletion downstream of the cut site and a single mismatch; these modifications (highlighted in gray) are flanked by 90-nt and 55-nt asymmetrical homology arms at 5′ and 3′ ends, respectively.
  • Template 2-7 (color coded) contain a HindIII and a BamHI sites that are flanked symmetrically by various lengths of homology arm, ranging from 30 nt to 250 nt.
  • Template 2-5 are synthetic single-stranded DNA oligonucleotides; whereas, template 6-7 are PCR amplified double-stranded DNA (see Materials and methods).
  • B HDR efficiency was tested in three cell conditions: unsynchronized, nocodazole synchronized, and nocodazole synchronized and aphidicolin blocked.
  • neoFb neonatal fibroblasts
  • hES human Embryonic Stem Cells
  • neoFb were subjected to a screening for the optimal cell cycle inhibitor with two doses of Cas9 RNP and template 4, and a different frequency profile was obtained ( FIG. 4A ).
  • nocodazole had no effect on neoFb cells.
  • Lovastatin, mimosine and thymidine continued to reduce the NHEJ and HDR frequencies, but hydroxyurea was no longer inhibitory.
  • Aphidicolin was the only tested treatment that enhanced NHEJ and HDR.
  • the NHEJ frequency increased from 8% to 18% at 30 ⁇ mol and 20% to 36% at 100 ⁇ mol Cas9 RNP.
  • the HDR frequency also increased from barely detectable to 2% at 30 ⁇ mol Cas9; at 100 ⁇ mol, HDR remained at 4% and no increase was seen ( FIG. 4A ).
  • the neoFb results demonstrate that different cell types require different synchronization methods.
  • FIG. 4A-4B depict NHEJ and HDR efficiencies at EMX1 locus in human primary neonatal fibroblast and embryonic stem cells.
  • A Primary neonatal fibroblast was subjected to the same inhibitor screening as described in the HEK293T experiment. One hundred ⁇ mol of Cas9 RNP and 100 ⁇ mol of HDR template 4 were used in the nucleofection reaction; the control reaction (C) contained no HDR template. The mean % NHEJ and HDR and standard deviation (error bar) were calculated from three experiments.
  • B Three cell conditions were tested in human ES cells: unsynchronized, nocodazole synchronized and nocodazole-aphidicolin sequential synchronized. Thirty or 100 pmol of Cas9 RNP was co-transfected with 100 ⁇ mol of HDR template 4, in the presence or absence of ROCK apoptosis inhibitor (10 ⁇ M).
  • Nocodazole synchronization enhanced the NHEJ frequency from 3-4% to 9% at 30 ⁇ mol and 10-14% to 28-31% at 100 ⁇ mol Cas9 RNP; however, no HDR event was detected ( FIG. 1 B).
  • the addition of ROCK apoptosis inhibitor improved the NHEJ frequency slightly.
  • a protocol from Pauklin et al 2013, Cell 155, 135-147) was modified: the cells were treated with nocodazole for 16 h, washed to remove the drug, and then treated with aphidicolin for 3 h before nucleofection.
  • a simple nocodazole treatment can achieve higher HDR efficiency at reduced dosage of Cas9 RNP.
  • Nocodazole blocks cells at M phase when the DNA is fully replicated and the nuclear membrane is broken down. Delivery of Cas9 RNP into a nocodazole synchronized cell may effectively target two cells because they divide upon release. Because the nuclear envelope is broken down, Cas9 RNP may efficiently gain access to the DNA.
  • Enhanced HDR frequency is useful for generating scar-less genetic tools (no introduction of drug selection gene), including epitope-tagged alleles, reporter genes, precise insertion, deletion and point mutations.
  • FIG. 6A Experimental timeline.
  • FIG. 6B Reporter system schematic.
  • a GFP gene (GFPA35) was mutated by insertion of a 35 bp fragment and integrated into human embryonic kidney (HEK) 293T and U2OS cells using lentivirus.
  • a targeting vector (t37GFP) containing a 5′ truncated GFP coding sequence was packaged into a recombinant AAV vector lacking a promoter. Homologous recombination between donor vector and defective GFP ⁇ 35 led to gene correction and GFP fluorescence.
  • FIG. 6C - FIG. 6F Percentage of GFP positive ( FIG. 6C , FIG. 6D ) HEK293T and ( FIG. 6E , FIG. 6F ) U2OS cells after treatment with 1 ⁇ M nocodazole and infection with ( FIG. 6C , FIG. 6E ) increasing multiplicity of infection (MOI) of AAV or ( FIG. 6D , FIG. 6F ) 1 ⁇ 10 5 MOI of AAV in the ( FIG. 6C , FIG. 6E ) absence or ( FIG. 6D , FIG. 6F ) presence of 1 ⁇ M Src7 (an inhibitor of nonhomologous end joining).
US15/502,720 2014-09-02 2015-09-01 Methods and compositions for RNA-directed target DNA modification Active US10570418B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/502,720 US10570418B2 (en) 2014-09-02 2015-09-01 Methods and compositions for RNA-directed target DNA modification

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462044812P 2014-09-02 2014-09-02
PCT/US2015/047966 WO2016036754A1 (fr) 2014-09-02 2015-09-01 Procédés et compositions pour assurer la modification de l'adn cible arn dépendante
US15/502,720 US10570418B2 (en) 2014-09-02 2015-09-01 Methods and compositions for RNA-directed target DNA modification

Publications (2)

Publication Number Publication Date
US20180044700A1 US20180044700A1 (en) 2018-02-15
US10570418B2 true US10570418B2 (en) 2020-02-25

Family

ID=55440319

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/502,720 Active US10570418B2 (en) 2014-09-02 2015-09-01 Methods and compositions for RNA-directed target DNA modification

Country Status (3)

Country Link
US (1) US10570418B2 (fr)
EP (1) EP3188763B1 (fr)
WO (1) WO2016036754A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022256448A2 (fr) 2021-06-01 2022-12-08 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes
WO2022266538A2 (fr) 2021-06-18 2022-12-22 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes humains
US11642374B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
WO2023167882A1 (fr) 2022-03-01 2023-09-07 Artisan Development Labs, Inc. Composition et méthodes d'insertion de transgène
WO2023225410A2 (fr) 2022-05-20 2023-11-23 Artisan Development Labs, Inc. Systèmes et procédés d'évaluation du risque d'événements d'édition génomique
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
KR101656237B1 (ko) * 2012-10-23 2016-09-12 주식회사 툴젠 표적 DNA에 특이적인 가이드 RNA 및 Cas 단백질을 암호화하는 핵산 또는 Cas 단백질을 포함하는, 표적 DNA를 절단하기 위한 조성물 및 이의 용도
BR112015026197B1 (pt) 2013-04-16 2022-12-06 Regeneron Pharmaceuticals, Inc Método para modificação marcada de um lócus genômico de interesse em uma célula de rato pluripotente
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
RU2725520C2 (ru) 2013-12-11 2020-07-02 Регенерон Фармасьютикалс, Инк. Способы и композиции для направленной модификации генома
US20150165054A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting caspase-9 point mutations
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
CA2963820A1 (fr) 2014-11-07 2016-05-12 Editas Medicine, Inc. Procedes pour ameliorer l'edition genomique mediee par crispr/cas
KR20230070319A (ko) 2014-11-21 2023-05-22 리제너론 파마슈티칼스 인코포레이티드 쌍 형성된 가이드 rna를 사용하는 표적화된 유전자 변형을 위한 방법 및 조성물
EP3283635B1 (fr) 2015-04-13 2022-07-06 Maxcyte, Inc. Procédés et compositions permettant de modifier l'adn génomique
EP3786294A1 (fr) 2015-09-24 2021-03-03 Editas Medicine, Inc. Utilisation d'exonucléases pour améliorer l'édition de génome à médiation par crispr/cas
WO2017070633A2 (fr) 2015-10-23 2017-04-27 President And Fellows Of Harvard College Protéines cas9 évoluées pour l'édition génétique
WO2017165826A1 (fr) 2016-03-25 2017-09-28 Editas Medicine, Inc. Systèmes d'édition de génome comprenant des molécules d'enzyme modulant la réparation et leurs procédés d'utilisation
WO2017180694A1 (fr) 2016-04-13 2017-10-19 Editas Medicine, Inc. Systèmes d'édition de gènes de molécules de fusion cas9 et leurs procédés d'utilisation
KR20240016444A (ko) 2016-05-20 2024-02-06 리제너론 파마슈티칼스 인코포레이티드 다중 가이드 RNAs를 이용한 면역학적 내성 파괴 방법
CN109983124B (zh) * 2016-06-02 2022-09-16 西格马-奥尔德里奇有限责任公司 使用可编程dna结合蛋白增强靶向基因组修饰
JP2019520844A (ja) * 2016-07-21 2019-07-25 マックスサイト インコーポレーティッド ゲノムdnaを改変するための方法および組成物
SG11201900907YA (en) 2016-08-03 2019-02-27 Harvard College Adenosine nucleobase editors and uses thereof
EP3497214B1 (fr) 2016-08-09 2023-06-28 President and Fellows of Harvard College Protéines de fusion cas9-recombinase programmables et utilisations associées
EP3500271A4 (fr) * 2016-08-19 2020-05-13 Bluebird Bio, Inc. Activateurs d'édition du génome
WO2018039438A1 (fr) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation d'acides aminés non naturels dans des protéines au moyen de l'édition de bases
CA3039928A1 (fr) 2016-10-14 2018-04-19 President And Fellows Of Harvard College Administration d'aav d'editeurs de nucleobases
WO2018119359A1 (fr) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Édition du gène récepteur ccr5 pour protéger contre l'infection par le vih
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
IL306092A (en) 2017-03-23 2023-11-01 Harvard College Nucleic base editors that include nucleic acid programmable DNA binding proteins
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
EP3638317A4 (fr) 2017-06-15 2021-03-17 The Regents of The University of California Insertions ciblées d'adn non viral
CN111183226A (zh) * 2017-07-11 2020-05-19 西格马-奥尔德里奇有限责任公司 使用核小体相互作用蛋白结构域来增强靶向基因组修饰
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11649442B2 (en) 2017-09-08 2023-05-16 The Regents Of The University Of California RNA-guided endonuclease fusion polypeptides and methods of use thereof
WO2019075373A1 (fr) * 2017-10-12 2019-04-18 Mayo Foundation For Medical Education And Research Procédés et compositions pour l'édition génique
WO2019079347A1 (fr) 2017-10-16 2019-04-25 The Broad Institute, Inc. Utilisations d'éditeurs de bases adénosine
MX2020004325A (es) 2017-10-27 2020-11-09 Univ California Reemplazo dirigido de receptores de células t endógenos.
US11661599B1 (en) 2017-12-14 2023-05-30 National Technology & Engineering Solutions Of Sandia, Llc CRISPR-Cas based system for targeting single-stranded sequences
AU2020242032A1 (en) 2019-03-19 2021-10-07 Massachusetts Institute Of Technology Methods and compositions for editing nucleotide sequences
US20210062248A1 (en) * 2019-08-26 2021-03-04 Pact Pharma Inc. Methods of performing guide-seq on primary human t cells
JP2022550599A (ja) 2019-10-03 2022-12-02 アーティサン ディベロップメント ラブズ インコーポレイテッド 操作されたデュアルガイド核酸を有するcrisprシステム
KR20230003467A (ko) 2020-03-19 2023-01-06 인텔리아 테라퓨틱스, 인크. 지시된 게놈 편집을 위한 방법 및 조성물
KR20230019843A (ko) 2020-05-08 2023-02-09 더 브로드 인스티튜트, 인코퍼레이티드 표적 이중 가닥 뉴클레오티드 서열의 두 가닥의 동시 편집을 위한 방법 및 조성물
IL308806A (en) 2021-06-01 2024-01-01 Arbor Biotechnologies Inc Gene editing systems including nuclease crisper and their uses
WO2024062138A1 (fr) 2022-09-23 2024-03-28 Mnemo Therapeutics Cellules immunitaires comprenant un gène suv39h1 modifié

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001019182A1 (fr) 1999-09-14 2001-03-22 David Wells Transfert nucleaire utilisant des cellules donneuses selectionnees
US20050207633A1 (en) 2003-04-02 2005-09-22 Nick Arini Method of, and computer software for, classification of cells into subpopulations
WO2005108622A2 (fr) * 2004-05-04 2005-11-17 University Of Delaware Methodes et kits permettant d'accroitre l'efficacite d'une alteration de sequences d'acides nucleiques dirigee a l'aide d'oligonucleotides
US20050273870A1 (en) 1999-10-14 2005-12-08 University Of Massachusetts Preparation and selection of donor cells for nuclear transplantation
WO2013176772A1 (fr) 2012-05-25 2013-11-28 The Regents Of The University Of California Procédés et compositions permettant la modification de l'adn cible dirigée par l'arn et la modulation de la transcription dirigée par l'arn
US8696359B2 (en) 2004-07-19 2014-04-15 Cambridge University Technical Services Limited Methods of assessing cognitive dysfunction
US20140227787A1 (en) 2012-12-12 2014-08-14 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US20140242699A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7153684B1 (en) 1992-10-08 2006-12-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
AU729377B2 (en) 1997-10-23 2001-02-01 Asterias Biotherapeutics, Inc. Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
US7410798B2 (en) 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
US20090227032A1 (en) 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
ES2367525T3 (es) 2005-12-13 2011-11-04 Kyoto University Factor de reprogramación celular.
JP2008307007A (ja) 2007-06-15 2008-12-25 Bayer Schering Pharma Ag 出生後のヒト組織由来未分化幹細胞から誘導したヒト多能性幹細胞
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
WO2010075303A1 (fr) 2008-12-23 2010-07-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Facteurs d'épissage avec un domaine de liaison à l'arn de protéine puf et domaine effecteur d'épissage et leurs utilisations
US9580714B2 (en) 2010-11-24 2017-02-28 The University Of Western Australia Peptides for the specific binding of RNA targets
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
WO2013141680A1 (fr) 2012-03-20 2013-09-26 Vilnius University Clivage d'adn dirigé par arn par le complexe cas9-arncr
EP2838912A4 (fr) 2012-04-16 2015-11-18 Univ Western Australia Peptides pour la liaison de cibles nucléotidiques
KR101656237B1 (ko) 2012-10-23 2016-09-12 주식회사 툴젠 표적 DNA에 특이적인 가이드 RNA 및 Cas 단백질을 암호화하는 핵산 또는 Cas 단백질을 포함하는, 표적 DNA를 절단하기 위한 조성물 및 이의 용도
PL3138910T3 (pl) 2012-12-06 2018-01-31 Sigma Aldrich Co Llc Oparta na CRISPR modyfikacja i regulacja genomu
PL2931898T3 (pl) 2012-12-12 2016-09-30 Le Cong Projektowanie i optymalizacja systemów, sposoby i kompozycje do manipulacji sekwencją z domenami funkcjonalnymi
EP2940140B1 (fr) 2012-12-12 2019-03-27 The Broad Institute, Inc. Ingénierie de systèmes, procédés et compositions de guidage optimisé pour manipulation de séquence
WO2014093701A1 (fr) 2012-12-12 2014-06-19 The Broad Institute, Inc. Génomique fonctionnelle employant des systèmes crispr-cas, des compositions, des procédés, des banques d'inactivation et leurs applications
WO2014093694A1 (fr) 2012-12-12 2014-06-19 The Broad Institute, Inc. Systèmes, procédés et compositions de crispr-nickase cas pour la manipulation de séquences dans les eucaryotes
EP3825401A1 (fr) 2012-12-12 2021-05-26 The Broad Institute, Inc. Systèmes de composants crispr-cas, procédés et compositions pour la manipulation de séquence
KR20150105634A (ko) 2012-12-12 2015-09-17 더 브로드 인스티튜트, 인코퍼레이티드 서열 조작을 위한 개선된 시스템, 방법 및 효소 조성물의 유전자 조작 및 최적화
PT2784162E (pt) 2012-12-12 2015-08-27 Broad Inst Inc Engenharia de sistemas, métodos e composições guia otimizadas para a manipulação de sequências
MX2015007743A (es) 2012-12-17 2015-12-07 Harvard College Ingenieria del genoma humano guiada por ácido robonucleico.
WO2014104878A1 (fr) 2012-12-27 2014-07-03 Keygene N.V. Procédé de suppression d'une liaison génétique dans un végétal
WO2014127287A1 (fr) * 2013-02-14 2014-08-21 Massachusetts Institute Of Technology Procédé pour la mutagenèse ciblée in vivo
US20150165054A1 (en) * 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting caspase-9 point mutations

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001019182A1 (fr) 1999-09-14 2001-03-22 David Wells Transfert nucleaire utilisant des cellules donneuses selectionnees
US20050273870A1 (en) 1999-10-14 2005-12-08 University Of Massachusetts Preparation and selection of donor cells for nuclear transplantation
US20050207633A1 (en) 2003-04-02 2005-09-22 Nick Arini Method of, and computer software for, classification of cells into subpopulations
WO2005108622A2 (fr) * 2004-05-04 2005-11-17 University Of Delaware Methodes et kits permettant d'accroitre l'efficacite d'une alteration de sequences d'acides nucleiques dirigee a l'aide d'oligonucleotides
US8696359B2 (en) 2004-07-19 2014-04-15 Cambridge University Technical Services Limited Methods of assessing cognitive dysfunction
WO2013176772A1 (fr) 2012-05-25 2013-11-28 The Regents Of The University Of California Procédés et compositions permettant la modification de l'adn cible dirigée par l'arn et la modulation de la transcription dirigée par l'arn
US20140227787A1 (en) 2012-12-12 2014-08-14 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US20140242699A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
Glucksmann et al., 2015, US 20150232881 A1, effective filing date, Nov. 7, 2013. *
Heyer, et al.; "Regulation of Homologous Recombination in Eukaryotes"; Annu. Rev. Genet.; vol. 44, pp. 113-139 (2010).
Kaur et al., 2009, Current Gene Therapy, vol. 9. p. 434-458. *
Kotterman et al., 2014, Nature Reviews, vol. 15, p. 445-451. *
Krouse et al., 2012, US 20120088807 A1. *
Lenzi et al., 2014, NCBI Bookshelf, A Service of the National Library of Medicine, National Institute of Health, Oversight and Review of Clinical Gene Transfer Protocols: Assessing the Role of the Recombinant DNA Advisory Committee. Washington (DC): National Academies Press (US), pp. 1-16. *
Lin, et al.; "Enhanced homology-directed human genome engineering by conrolled timing of CRISPR/Cas9 delivery"; eLife; 13 pages (Dec. 15, 2014).
Lundin, et al.; "Different Roles for Nonhomologous End Joining and Homologous Recombination following Replication Arrest in Mammalian Cells"; Molecular and Cellular Biology; vol. 22, No. 16, pp. 5869-5878 (Aug. 2002).
Orthwein, et al.; "Mitosis Inhibits DNA Double-Strand Break Repair to Guard Against Telomere Fusions"; Science; vol. 344, No. 6180 pp. 189-193 (Mar. 20, 2014).
Shrivastav, et al.; "Regulation of DNA double-strand break repair pathway choice"; Cell Research; vol. 18, pp. 134-147 (2008).

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11642374B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11642375B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11903966B2 (en) 2015-07-31 2024-02-20 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US11925664B2 (en) * 2015-07-31 2024-03-12 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
WO2022256448A2 (fr) 2021-06-01 2022-12-08 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes
WO2022266538A2 (fr) 2021-06-18 2022-12-22 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes humains
WO2023167882A1 (fr) 2022-03-01 2023-09-07 Artisan Development Labs, Inc. Composition et méthodes d'insertion de transgène
WO2023225410A2 (fr) 2022-05-20 2023-11-23 Artisan Development Labs, Inc. Systèmes et procédés d'évaluation du risque d'événements d'édition génomique

Also Published As

Publication number Publication date
WO2016036754A1 (fr) 2016-03-10
EP3188763B1 (fr) 2020-05-13
EP3188763A4 (fr) 2018-03-14
US20180044700A1 (en) 2018-02-15
EP3188763A1 (fr) 2017-07-12

Similar Documents

Publication Publication Date Title
US10570418B2 (en) Methods and compositions for RNA-directed target DNA modification
US10494620B2 (en) Methods and compositions for modifying a single stranded target nucleic acid
US11180792B2 (en) Methods and compositions for labeling a single-stranded target nucleic acid
US11085057B2 (en) Compositions and methods for modifying a target nucleic acid
US10392607B2 (en) Cas9 variants and methods of use thereof
EP3307762B1 (fr) Variants de cas9 rapporteurs et leurs procédés d'utilisation
US10253311B2 (en) Methods and compositions for using argonaute to modify a single stranded target nucleic acid
JP2018532402A (ja) Rnaプログラム可能エンドヌクレアーゼの新規のファミリーならびにゲノム編集および他の適用におけるそれらの使用
US20220145274A1 (en) Novel high fidelity rna-programmable endonuclease systems and uses thereof
US20240141312A1 (en) Type v rna programmable endonuclease systems
EP4101928A1 (fr) Systèmes d'endonucléase programmables à arn de type v
WO2023118068A1 (fr) Nouveaux petits systèmes programmables d'endonucléases à arn de type v
WO2023237587A1 (fr) Nouveaux petits systèmes programmables d'endonucléases à arn de type v
WO2023036669A1 (fr) Nouveaux systèmes d'endonucléases programmables à petits arn à spécificité pam améliorée et leurs utilisations

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOUDNA, JENNIFER A.;LIN, STEVEN;STAAHL, BRETT T.;SIGNING DATES FROM 20150904 TO 20170411;REEL/FRAME:042258/0240

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOUDNA, JENNIFER A.;LIN, STEVEN;STAAHL, BRETT T.;SIGNING DATES FROM 20150904 TO 20170411;REEL/FRAME:042258/0240

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4